193 results on '"McCrae KR"'
Search Results
2. Annexin A2: biology and relevance to the antiphospholipid syndrome
- Author
-
Cockrell, E, primary, Espinola, RG, additional, and McCrae, KR, additional
- Published
- 2008
- Full Text
- View/download PDF
3. Antiphospholipid antibody associated thrombosis: A consensus for treatment?
- Author
-
McCrae, KR, primary
- Published
- 1996
- Full Text
- View/download PDF
4. Interaction of single-chain urokinase with its receptor induces the appearance and disappearance of binding epitopes within the resultant complex for other cell surface proteins
- Author
-
Higazi, AA, primary, Upson, RH, additional, Cohen, RL, additional, Manuppello, J, additional, Bognacki, J, additional, Henkin, J, additional, McCrae, KR, additional, Kounnas, MZ, additional, Strickland, DK, additional, Preissner, KT, additional, Lawler, J, additional, and Cines, DB, additional
- Published
- 1996
- Full Text
- View/download PDF
5. Detection of antitrophoblast antibodies in the sera of patients with anticardiolipin antibodies and fetal loss
- Author
-
McCrae, KR, primary, DeMichele, AM, additional, Pandhi, P, additional, Balsai, MJ, additional, Samuels, P, additional, Graham, C, additional, Lala, PK, additional, and Cines, DB, additional
- Published
- 1993
- Full Text
- View/download PDF
6. Pregnancy-associated thrombocytopenia: pathogenesis and management
- Author
-
McCrae, KR, primary, Samuels, P, additional, and Schreiber, AD, additional
- Published
- 1992
- Full Text
- View/download PDF
7. Characterization of urokinase receptor expression by human placental trophoblasts
- Author
-
Zini, JM, primary, Murray, SC, additional, Graham, CH, additional, Lala, PK, additional, Kariko, K, additional, Barnathan, ES, additional, Mazar, A, additional, Henkin, J, additional, Cines, DB, additional, and McCrae, KR, additional
- Published
- 1992
- Full Text
- View/download PDF
8. beta(2)-glycoprotein i is a cofactor for tissue plasminogen activator-mediated plasminogen activation.
- Author
-
Bu C, Gao L, Xie W, Zhang J, He Y, Cai G, and McCrae KR
- Abstract
OBJECTIVE: Regulation of the conversion of plasminogen to plasmin by tissue plasminogen activator (tPA) is critical in the control of fibrin deposition. While several plasminogen activators have been described, soluble plasma cofactors that stimulate fibrinolysis have not been characterized. The purpose of this study was to investigate the effects of beta(2)-glycoprotein I (beta(2)GPI), an abundant plasma glycoprotein, on tPA-mediated plasminogen activation. METHODS: The effect of beta(2)GPI on tPA-mediated activation of plasminogen was assessed using amidolytic assays, a fibrin gel, and plasma clots. Binding of beta(2)GPI to tPA and plasminogen was determined in parallel. The effects of IgG fractions and anti-beta(2)GPI antibodies from patients with antiphospholipid syndrome (APS) on tPA-mediated plasminogen activation were also measured. RESULTS: Beta(2)-glycoprotein I stimulated tPA-dependent plasminogen activation in the fluid phase and within a fibrin gel. The beta(2)GPI region responsible for stimulating tPA activity was shown to be at least partly contained within beta(2)GPI domain V. In addition, beta(2)GPI bound tPA with high affinity (K(d) approximately 20 nM), stimulated tPA amidolytic activity, and caused an overall 20-fold increase in the catalytic efficiency (K(cat)/K(m)) of tPA-mediated conversion of Glu-plasminogen to plasmin. Moreover, depletion of beta(2)GPI from plasma led to diminished rates of clot lysis, with restoration of normal lysis rates following beta(2)GPI repletion. Stimulation of tPA-mediated plasminogen activity by beta(2)GPI was inhibited by monoclonal anti-beta(2)GPI antibodies as well as by anti-beta(2)GPI antibodies from patients with APS. CONCLUSION: These findings suggest that beta(2)GPI may be an endogenous regulator of fibrinolysis. Impairment of beta(2)GPI-stimulated fibrinolysis by anti-beta(2)GPI antibodies may contribute to the development of thrombosis in patients with APS. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
9. Immune thrombocytopenic purpura in pregnancy.
- Author
-
Gernsheimer T, McCrae KR, Gernsheimer, Terry, and McCrae, Keith R
- Published
- 2007
- Full Text
- View/download PDF
10. Endothelial cells in physiology and in the pathophysiology of vascular disorders
- Author
-
Cines, Db, Pollak, Es, Buck, Ca, Loscalzo, J., Zimmerman, Ga, Mcever, Rp, Pober, Js, Wick, Tm, Barbara Konkle, Schwartz, Bs, Barnathan, Es, Mccrae, Kr, Hug, Ba, Schmidt, Am, and Stern, Dm
11. Complement Biosensors Identify a Classical Pathway Stimulus in Complement-Mediated Thrombotic Microangiopathy.
- Author
-
Cole MA, Ranjan N, Gerber GF, Pan XZ, Flores-Guerrero D, McNamara G, Chaturvedi S, Sperati CJ, McCrae KR, and Brodsky RA
- Abstract
Complement-mediated thrombotic microangiopathy or hemolytic uremic syndrome (CM-TMA/CM-HUS), previously identified as atypical hemolytic uremic syndrome, is a thrombotic microangiopathy characterized by germline variants or acquired antibodies to complement proteins and regulators. Building upon our prior experience with the modified Ham (mHam) assay for ex vivo diagnosis of complementopathies, we have developed an array of cell-based complement "biosensors' by selective removal of complement regulatory proteins (CD55 and CD59, CD46, or a combination thereof) in an autonomously bioluminescent HEK293 cell line. These biosensors can be used as a sensitive method for diagnosing CM-TMA and monitoring therapeutic complement blockade. Using specific complement pathway inhibitors, this model identifies IgM-driven classical pathway stimulus during both acute disease and in many patients during clinical remission. This provides a potential explanation for ~50% of CM-TMA patients who lack an alternative pathway "driving" variant and suggests at least a subset of CM-TMA is characterized by a breakdown of IgM immunologic tolerance., (Copyright © 2024 American Society of Hematology.)
- Published
- 2024
- Full Text
- View/download PDF
12. Pomalidomide for Epistaxis in Hereditary Hemorrhagic Telangiectasia.
- Author
-
Al-Samkari H, Kasthuri RS, Iyer VN, Pishko AM, Decker JE, Weiss CR, Whitehead KJ, Conrad MB, Zumberg MS, Zhou JY, Parambil J, Marsh D, Clancy M, Bradley L, Wisniewski L, Carper BA, Thomas SM, and McCrae KR
- Subjects
- Aged, Female, Humans, Male, Middle Aged, Angiogenesis Inhibitors administration & dosage, Angiogenesis Inhibitors adverse effects, Double-Blind Method, Quality of Life, Severity of Illness Index, Treatment Outcome, Neutropenia chemically induced, Neutropenia epidemiology, Constipation chemically induced, Constipation epidemiology, Drug Eruptions epidemiology, Drug Eruptions etiology, Epistaxis diagnosis, Epistaxis drug therapy, Epistaxis etiology, Epistaxis psychology, Telangiectasia, Hereditary Hemorrhagic complications, Telangiectasia, Hereditary Hemorrhagic drug therapy, Thalidomide administration & dosage, Thalidomide adverse effects, Thalidomide analogs & derivatives
- Abstract
Background: Hereditary hemorrhagic telangiectasia (HHT) is characterized by extensive telangiectasias and arteriovenous malformations. The primary clinical manifestation is epistaxis that results in iron-deficiency anemia and reduced health-related quality of life., Methods: We conducted a randomized, placebo-controlled trial to evaluate the safety and efficacy of pomalidomide for the treatment of HHT. We randomly assigned patients, in a 2:1 ratio, to receive pomalidomide at a dose of 4 mg daily or matching placebo for 24 weeks. The primary outcome was the change from baseline through week 24 in the Epistaxis Severity Score (a validated bleeding score in HHT; range, 0 to 10, with higher scores indicating worse bleeding). A reduction of 0.71 points or more is considered clinically significant. A key secondary outcome was the HHT-specific quality-of-life score (range, 0 to 16, with higher scores indicating more limitations)., Results: The trial was closed to enrollment in June 2023 after a planned interim analysis met a prespecified threshold for efficacy. A total of 144 patients underwent randomization; 95 patients were assigned to receive pomalidomide and 49 to receive placebo. The baseline mean (±SD) Epistaxis Severity Score was 5.0±1.5, a finding consistent with moderate-to-severe epistaxis. At 24 weeks, the mean difference between the pomalidomide group and the placebo group in the change from baseline in the Epistaxis Severity Score was -0.94 points (95% confidence interval [CI], -1.57 to -0.31; P = 0.004). The mean difference in the changes in the HHT-specific quality-of-life score between the groups was -1.4 points (95% CI, -2.6 to -0.3). Adverse events that were more common in the pomalidomide group than in the placebo group included neutropenia, constipation, and rash., Conclusions: Among patients with HHT, pomalidomide treatment resulted in a significant, clinically relevant reduction in epistaxis severity. No unexpected safety signals were identified. (Funded by the National Heart, Lung, and Blood Institute; PATH-HHT Clinicaltrials.gov number, NCT03910244)., (Copyright © 2024 Massachusetts Medical Society.)
- Published
- 2024
- Full Text
- View/download PDF
13. Factor XII promotes the thromboinflammatory response in a rat model of venoarterial extracorporeal membrane oxygenation.
- Author
-
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, and Palumbo JS
- Subjects
- Animals, Male, Rats, Anticoagulants pharmacology, Arginine analogs & derivatives, Blood Coagulation drug effects, Disease Models, Animal, Factor XII Deficiency genetics, Inflammation metabolism, Inflammation genetics, Pipecolic Acids pharmacology, Rats, Sprague-Dawley, Sulfonamides pharmacology, Extracorporeal Membrane Oxygenation, Factor XII genetics, Factor XII metabolism, Thrombosis etiology, Thrombosis genetics
- 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., (Copyright © 2023 The American Association for Thoracic Surgery. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
14. The 2022 review of the 2019 American Society of Hematology guidelines on immune thrombocytopenia.
- Author
-
Neunert CE, Arnold DM, Grace RF, Kuhne T, McCrae KR, and Terrell DR
- Subjects
- Adult, Humans, Disease Management, Societies, Medical, United States, Hematology standards, Practice Guidelines as Topic, Purpura, Thrombocytopenic, Idiopathic therapy, Purpura, Thrombocytopenic, Idiopathic diagnosis
- Abstract
Abstract: The 2019 American Society of Hematology (ASH) guidelines for immune thrombocytopenia (ITP) included recommendations on the management of adults (recommendations 1-9) and children (recommendations 10-21) with primary ITP . We describe here the results of a review of the 2019 guidelines by a working group of experts requested by ASH to inform decision-making about the need for and timing of a guideline revision. An updated Medline and Embase search applied the same search terms as in the 2019 ASH guidelines, limited to systematic reviews and clinical trials, from May 2017 to July 2022. There were 193 studies identified, 102 underwent abstract reviews, and 54 full reviews. Each study was assessed based on relevance to the previous recommendation with regard to the population, prioritized outcomes, new outcomes, and study design. Reviewers assessed if the data would change the strength or the directionality of the existing recommendation or merit development of a new recommendation. Based on this review, the ASH Committee on Quality endorsed a focused update on second-line management for adults with ITP. In addition, there will be continued annual monitoring and reviewing of the 2019 ASH guidelines on ITP in full to evaluate when there is sufficient new evidence to warrant additional revisions., (© 2024 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
15. Catastrophic antiphospholipid syndrome: challenging case and importance of multidisciplinary evaluation and management.
- Author
-
Sorathia S, Arockiam AD, Haroun E, Khurana R, Hall A, McCarthy M, Shastri RK, Sawaf H, McCrae KR, Jellis CL, and Wang TKM
- Abstract
Our case depicts a challenging diagnosis of catastrophic antiphospholipid syndrome in a young patient with a heterogenous presentation with extensive clinical course, a wide range of investigations, including multimodality imaging, and multidisciplinary expertise, to initiate prompt treatment addressing multiorgan thrombotic injury., Competing Interests: No conflicts of interest for all authors., (© 2024 The Author(s). Clinical Case Reports published by John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
16. Complement Biosensors Identify a Classical Pathway Stimulus in Complement-Mediated Hemolytic Uremic Syndrome.
- Author
-
Cole MA, Ranjan N, Gerber GF, Pan XZ, Flores-Guerrero D, Chaturvedi S, Sperati CJ, McCrae KR, and Brodsky RA
- Abstract
Complement-mediated hemolytic uremic syndrome (CM-HUS) is a thrombotic microangiopathy characterized by germline variants or acquired antibodies to complement proteins and regulators. Building upon our prior experience with the modified Ham (mHam) assay for ex vivo diagnosis of complementopathies, we have developed an array of cell-based complement "biosensors'' by selective removal of complement regulatory proteins (CD55 and CD59, CD46, or a combination thereof) in an autonomously bioluminescent HEK293 cell line. These biosensors can be used as a sensitive method for diagnosing CM-HUS and monitoring therapeutic complement blockade. Using specific complement pathway inhibitors, this model identifies IgM-driven classical pathway stimulus during both acute disease and in many patients during clinical remission. This provides a potential explanation for ~50% of CM-HUS patients who lack an alternative pathway "driving" variant and suggests at least a subset of CM-HUS is characterized by a breakdown of IgM immunologic tolerance., Key Points: CM-HUS has a CP stimulus driven by polyreactive IgM, addressing the mystery of why 40% of CM-HUS lack complement specific variantsComplement biosensors and the bioluminescent mHam can be used to aid in diagnosis of CM-HUS and monitor complement inhibitor therapy.
- Published
- 2024
- Full Text
- View/download PDF
17. A "backup plan" for ADAMTS13 deficiency in TTP.
- Author
-
Hubben A and McCrae KR
- Subjects
- Humans, Mutation, ADAMTS13 Protein deficiency, ADAMTS13 Protein metabolism, ADAMTS13 Protein genetics, Purpura, Thrombotic Thrombocytopenic genetics, Purpura, Thrombotic Thrombocytopenic diagnosis, Purpura, Thrombotic Thrombocytopenic blood
- Published
- 2024
- Full Text
- View/download PDF
18. Dysregulated platelet function in patients with postacute sequelae of COVID-19.
- Author
-
Aggarwal A, Singh TK, Pham M, Godwin M, Chen R, McIntyre TM, Scalise A, Chung MK, Jennings C, Ali M, Park H, Englund K, Khorana AA, Svensson LG, Kapadia S, McCrae KR, and Cameron SJ
- Subjects
- Humans, SARS-CoV-2, Factor Xa, Blood Coagulation, Disease Progression, COVID-19 complications, Thrombosis etiology
- Abstract
Background: Postacute sequelae of COVID-19 (PASC), also referred to as "Long COVID", sometimes follows COVID-19, a disease caused by SARS-CoV-2. Although SARS-CoV-2 is well known to promote a prothrombotic state, less is known about the thrombosis risk in PASC. Our objective was to evaluate platelet function and thrombotic potential in patients following recovery from SARS-CoV-2, but with clear symptoms of patients with PASC., Methods: patients with PASC and matched healthy controls were enrolled in the study on average 15 months after documented SARS-CoV-2 infection. Platelet activation was evaluated by light transmission aggregometry (LTA) and flow cytometry in response to platelet surface receptor agonists. Thrombosis in platelet-deplete plasma was evaluated by Factor Xa activity. A microfluidics system assessed thrombosis in whole blood under shear stress conditions., Results: A mild increase in platelet aggregation in patients with PASC through the thromboxane receptor was observed, and platelet activation through the glycoprotein VI (GPVI) receptor was decreased in patients with PASC compared to age- and sex-matched healthy controls. Thrombosis under shear conditions as well as Factor Xa activity were reduced in patients with PASC. Plasma from patients with PASC was an extremely potent activator of washed, healthy platelets - a phenomenon not observed when stimulating healthy platelets after incubation with plasma from healthy individuals., Conclusions: patients with PASC show dysregulated responses in platelets and coagulation in plasma, likely caused by a circulating molecule that promotes thrombosis. A hitherto undescribed protective response appears to exist in patients with PASC to counterbalance ongoing thrombosis that is common to SARS-CoV-2 infection., Competing Interests: Declaration of conflicting interestsThe authors declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
- Published
- 2024
- Full Text
- View/download PDF
19. A machine learning approach to predict mortality due to immune-mediated thrombotic thrombocytopenic purpura.
- Author
-
Abou-Ismail MY, Zhang C, Presson AP, Chaturvedi S, Antun AG, Farland AM, Woods R, Metjian A, Park YA, de Ridder G, Gibson B, Kasthuri RS, Liles DK, Akwaa F, Clover T, Kreuziger LB, Sridharan M, Go RS, McCrae KR, Upreti HV, Gangaraju R, Kocher NK, Zheng XL, Raval JS, Masias C, Cataland SR, Johnson AD, Davis E, Evans MD, Mazepa M, and Lim MY
- Abstract
Background: Mortality due to immune-mediated thrombotic thrombocytopenic purpura (iTTP) remains significant. Predicting mortality risk may potentially help individualize treatment. The French Thrombotic Microangiopathy (TMA) Reference Score has not been externally validated in the United States. Recent advances in machine learning technology can help analyze large numbers of variables with complex interactions for the development of prediction models., Objectives: To validate the French TMA Reference Score in the United States Thrombotic Microangiopathy (USTMA) iTTP database and subsequently develop a novel mortality prediction tool, the USTMA TTP Mortality Index., Methods: We analyzed variables available at the time of initial presentation, including demographics, symptoms, and laboratory findings. We developed our model using gradient boosting machine, a machine learning ensemble method based on classification trees, implemented in the R package gbm., Results: In our cohort ( n = 419), the French score predicted mortality with an area under the receiver operating characteristic curve of 0.63 (95% CI: 0.50-0.77), sensitivity of 0.35, and specificity of 0.84. Our gradient boosting machine model selected 8 variables to predict acute mortality with a cross-validated area under the receiver operating characteristic curve of 0.77 (95% CI: 0.71-0.82). The 2 cutoffs corresponded to sensitivities of 0.64 and 0.50 and specificities of 0.76 and 0.87, respectively., Conclusion: The USTMA Mortality Index was acceptable for predicting mortality due to acute iTTP in the USTMA registry, but not sensitive enough to rule out death. Identifying patients at high risk of iTTP-related mortality may help individualize care and ultimately improve iTTP survival outcomes. Further studies are needed to provide external validation. Our model is one of many recent examples where machine learning models may show promise in clinical prediction tools in healthcare., (© 2024 The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
20. A descriptive analysis of fatal outcomes in immune thrombotic thrombocytopenic purpura in the USTMA TTP Registry.
- Author
-
Abou-Ismail MY, Zhang C, Presson AP, Chaturvedi S, Antun AG, Farland AM, Woods R, Metjian A, Park YA, de Ridder G, Gibson B, Kasthuri RS, Liles DK, Akwaa F, Clover T, Baumann Kreuziger L, Sridharan M, Go RS, McCrae KR, Upreti HV, Gangaraju R, Kocher NK, Zheng XL, Raval JS, Masias C, Cataland SR, Johnson AD, Davis E, Evans MD, Mazepa M, and Lim MY
- Subjects
- Humans, Registries, Purpura, Thrombotic Thrombocytopenic epidemiology, Purpura, Thrombocytopenic, Idiopathic, Thrombosis
- Published
- 2024
- Full Text
- View/download PDF
21. High molecular weight kininogen interactions with the homologs prekallikrein and factor XI: importance to surface-induced coagulation.
- Author
-
Mohammed BM, Sun MF, Cheng Q, Litvak M, McCrae KR, Emsley J, McCarty OJT, and Gailani D
- Subjects
- Animals, Humans, Mice, Factor XI metabolism, Prekallikrein metabolism, Blood Coagulation, Kininogen, High-Molecular-Weight metabolism, Thrombosis
- 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., Competing Interests: Declaration of competing interests D.G. is a consultant for pharmaceutical companies (Anthos Therapeutics; Aronora, Inc.; Bayer Pharma; Bristol-Myers Squibb; Ionis Pharmaceuticals; Janssen, Pharmaceuticals) with interests in targeting FXI, FXII, and prekallikrein for therapeutic purposes. The other authors have no conflicts to report., (Copyright © 2023 International Society on Thrombosis and Haemostasis. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
22. How to diagnose and manage antiphospholipid syndrome.
- Author
-
Hubben A and McCrae KR
- Subjects
- Female, Pregnancy, Humans, Antibodies, Antiphospholipid, Lupus Coagulation Inhibitor, beta 2-Glycoprotein I, Immunoglobulin A, Antiphospholipid Syndrome diagnosis, Antiphospholipid Syndrome therapy, Thrombosis diagnosis, Thrombosis therapy
- Abstract
Antiphospholipid antibodies (aPL) are autoimmune antibodies directed toward phospholipids or phospholipid-protein complexes, particularly those containing β2-glycoprotein I (β2GPI). Persistently positive aPL accompanied by arterial or venous thrombosis, or recurrent pregnancy loss, constitutes the antiphospholipid syndrome (APS). Several types of aPL with different specificities have been defined and may be detected in the clinical lab, including lupus anticoagulants (detected using clotting assays) and anticardiolipin, anti-β2GPI and anti-prothrombin/phosphatidylserine antibodies (detected by ELISA); each of the last 3 aPL may be either IgG, IgM, or IgA, though IgA antibodies are not included in criteria for APS. Due to the relative rarity of APS and the heterogeneity of aPL, thrombosis risk stratification is challenging, and randomized clinical trials for thrombosis treatment and prevention have been limited. This lack of high-quality data has made the clinical management of APS difficult, and existing guidelines are few and could not possibly cover many of the scenarios encountered in managing patients with APS. In this review, we present 3 patients with aPL and/or APS who highlight treatment dilemmas, and we discuss background information that may help guide clinical judgment in developing individualized treatment plans for patients with these enigmatic antibodies., (Copyright © 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
23. Dysregulated Platelet Function in Patients with Post-Acute Sequelae of COVID-19.
- Author
-
Aggarwal A, Singh TK, Pham M, Godwin M, Chen R, McIntyre TM, Scalise A, Chung MK, Jennings C, Ali M, Park H, Englund K, Khorana AA, Svensson LG, Kapadia S, McCrae KR, and Cameron SJ
- Abstract
Background: Post-acute sequelae of COVID-19 (PASC), also referred as Long-COVID, sometimes follows COVID-19, a disease caused by SARS-CoV-2. While SARS-CoV-2 is well-known to promote a prothrombotic state, less is known about the thrombosis risk in PASC., Aim: Our objective was to evaluate the platelet function and thrombotic potential in patients following recovery from SARS-CoV-2 with clear symptoms of PASC., Methods: PASC patients and matched healthy controls were enrolled in the study on average 15 months after documented SARS-CoV-2 infection. Platelet activation was evaluated by Light Transmission Aggregometry (LTA) and flow cytometry in response to platelet surface receptor agonists. Thrombosis in platelet-deplete plasma was evaluated by Factor Xa activity. A microfluidics system assessed thrombosis in whole blood under shear stress conditions., Results: A mild increase in platelet aggregation in PASC patients through the thromboxane receptor was observed and platelet activation through the glycoprotein VI (GPVI) receptor was decreased in PASC patients compared to age- and sex-matched healthy controls. Thrombosis under shear conditions as well as Factor Xa activity were reduced in PASC patients. Plasma from PASC patients was an extremely potent activator of washed, healthy platelets - a phenomenon not observed when stimulating healthy platelets after incubation with plasma from healthy individuals., Conclusions: PASC patients show dysregulated responses in platelets and coagulation in plasma, likely caused by a circulating molecule that promotes thrombosis. A hitherto undescribed protective response appears to exists in PASC patients to counterbalance ongoing thrombosis that is common to SARS-CoV-2 infection.
- Published
- 2023
- Full Text
- View/download PDF
24. Transcriptome analysis suggests a central role for complement and C5aR1 in neutrophil activation in APS.
- Author
-
Alarabi A, Hubben A, Barnard J, Knight JS, and McCrae KR
- Subjects
- Humans, Complement System Proteins, Gene Expression Profiling, Complement Activation, Neutrophil Activation, Antiphospholipid Syndrome
- Abstract
Competing Interests: Declaration of competing interest None of the authors have any financial or intellectual conflicts of interest.
- Published
- 2023
- Full Text
- View/download PDF
25. Protection of β2GPI Deficient Mice from Thrombosis Reflects a Defect in PAR3-facilitated Platelet Activation.
- Author
-
Kulkarni PP, Alluri RK, Godwin M, Forbes GL, Merkulova A, Vijay A, Palihati M, Kundu S, Jun-Shim Y, Schmaier A, Holinstat M, Cameron SJ, and McCrae KR
- Abstract
Background: Antibodies to β2-glycoprotein I (β2GPI) cause thrombosis in antiphospholipid syndrome, however the role of β2GPI itself in regulation of coagulation pathways in vivo is not well understood., Methods: We developed β2GPI-deficient mice (Apoh
-/- ) by deleting exon 2 and 3 of Apoh using CRISPR/Cas9 and compared the propensity of wild-type (WT) and Apoh-/- mice to develop thrombosis using rose bengal and FeCl3 -induced carotid thrombosis, laser-induced cremaster arteriolar injury, and inferior vena cava (IVC) stasis models. We also compared tail bleeding times and assessed platelet activation in WT and Apoh-/- mice in the absence and presence of exogenous β2GPI., Results: Compared to WT littermates, Apoh-/- mice demonstrated a prolonged time to occlusion of the carotid artery after exposure to rose bengal or FeCl3 , and reduced platelet and fibrin accumulation in cremasteric arterioles after laser injury. Similarly, significantly smaller thrombi were retrieved from the IVC of Apoh-/- mice 48 hours after IVC occlusion. The activated partial thromboplastin time (aPTT) and prothrombin time, as well as aPTT reagent- and tissue factor-induced thrombin generation times using plasma from Apoh-/- and WT mice revealed no differences. However, we observed significant prolongation of tail bleeding in Apoh-/- mice, and reduced P-selectin expression and binding of fibrinogen to the activated α2bβ3 integrin on platelets from these mice after stimulation with low thrombin concentrations; these changes were reversed by exogenous β2GPI. An antibody to PAR3 blocked thrombin-induced activation of WT, but not Apoh-/- platelets, as well as the ability of β2GPI to restore the activation response of Apoh-/- platelets to thrombin. β2GPI deficiency did not affect platelet activation by a PAR4-activator peptide, or ADP., Conclusions: In mice, β2GPI may mediate procoagulant activity by enhancing the ability of PAR3 to present thrombin to PAR4, promoting platelet activation at low thrombin concentrations., Key Points: β2GPI deficient mice are protected from experimental arterial, venous, and microvascular thrombosis.β2GPI deficient mice display prolonged tail bleeding times and reduced PAR3-facilitated platelet activation by low concentrations of thrombin.- Published
- 2023
- Full Text
- View/download PDF
26. C1 inhibitor deficiency enhances contact pathway-mediated activation of coagulation and venous thrombosis.
- Author
-
Grover SP, Kawano T, Wan J, Tanratana P, Polai Z, Shim YJ, Snir O, Brækkan S, Dhrolia S, Kasthuri RR, Bendapudi PK, McCrae KR, Wolberg AS, Hansen JB, Farkas H, and Mackman N
- Subjects
- Humans, Animals, Mice, Thrombin, Complement C1 Inhibitor Protein genetics, Blood Coagulation, Angioedemas, Hereditary genetics, Thrombosis etiology, Venous Thrombosis etiology
- Abstract
C1 inhibitor (C1INH) is a multifunctional serine protease inhibitor that functions as a major negative regulator of several biological pathways, including the contact pathway of blood coagulation. In humans, congenital C1INH deficiency results in a rare episodic bradykinin-mediated swelling disorder called hereditary angioedema (HAE). Patients with C1INH deficiency-associated HAE (C1INH-HAE) have increased circulating markers of activation of coagulation. Furthermore, we recently reported that patients with C1INH-HAE had a moderate but significant increased risk of venous thromboembolism. To further investigate the impact of C1INH deficiency on activation of coagulation and thrombosis, we conducted studies using patient samples and mouse models. Plasmas from patients with C1INH-HAE had significantly increased contact pathway-mediated thrombin generation. C1INH-deficient mice, which have been used as a model of C1INH-HAE, had significantly increased baseline circulating levels of prothrombin fragment 1+2 and thrombin-antithrombin complexes. In addition, whole blood from C1INH-deficient mice supported significantly increased contact pathway-mediated thrombin generation. Importantly, C1INH-deficient mice exhibited significantly enhanced venous, but not arterial, thrombus formation. Furthermore, purified human C1INH normalized contact pathway-mediated thrombin generation and venous thrombosis in C1INH-deficient mice. These findings highlight a key role for endogenous C1INH as a negative regulator of contact pathway-mediated coagulation in humans and mice. Further, this work identifies endogenous C1INH as an important negative regulator of venous thrombus formation in mice, complementing the phenotype associated with C1INH-HAE., (© 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
27. Anti-HK antibody inhibits the plasma contact system by blocking prekallikrein and factor XI activation in vivo.
- Author
-
Chen ZL, Singh PK, Horn K, Calvano MR, Kaneki S, McCrae KR, Strickland S, and Norris EH
- Subjects
- Humans, Animals, Mice, Factor XI metabolism, Bradykinin pharmacology, Bradykinin chemistry, Kininogen, High-Molecular-Weight chemistry, Kininogen, High-Molecular-Weight metabolism, Prekallikrein chemistry, Prekallikrein metabolism, Thrombosis
- Abstract
A dysregulated plasma contact system is involved in various pathological conditions, such as hereditary angioedema, Alzheimer disease, and sepsis. We previously showed that the 3E8 anti-high molecular weight kininogen (anti-HK) antibody blocks HK cleavage and bradykinin generation in human plasma ex vivo. Here, we show that 3E8 prevented not only HK cleavage but also factor XI (FXI) and prekallikrein (PK) activation by blocking their binding to HK in mouse plasma in vivo. 3E8 also inhibited contact system-induced bradykinin generation in vivo. Interestingly, FXII activation was also inhibited, likely because of the ability of 3E8 to block the positive feedback activation of FXII by kallikrein (PKa). In human plasma, 3E8 also blocked PK and FXI binding to HK and inhibited both thrombotic (FXI activation) and inflammatory pathways (PK activation and HK cleavage) of the plasma contact system activation ex vivo. Moreover, 3E8 blocked PKa binding to HK and dose-dependently inhibited PKa cleavage of HK. Our results reveal a novel strategy to inhibit contact system activation in vivo, which may provide an effective method to treat human diseases involving contact system dysregulation., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
28. Emerging Therapies in Antiphospholipid Syndrome.
- Author
-
Hubben A and McCrae KR
- Subjects
- Pregnancy, Female, Humans, Hydroxychloroquine therapeutic use, Rituximab therapeutic use, Antibodies, Antiphospholipid therapeutic use, Anticoagulants therapeutic use, Antiphospholipid Syndrome drug therapy, Thrombosis drug therapy, Thrombosis etiology, Thrombosis prevention & control
- Abstract
The antiphospholipid syndrome (APS) is the most common cause of acquired immune-mediated thrombophilia. This syndrome is broadly defined by the presence of arterial or venous thrombosis, or pregnancy morbidity, in the presence of high levels of antiphospholipid antibodies. Despite recognition of this disorder more than 50 years ago, a fundamental unifying pathogenesis has not been determined. Due to this, mechanism-based therapies for APS are not available, and current management following thrombotic events suggests anticoagulation of indeterminate duration, or for obstetric complications, heparin/low molecular weight heparin and aspirin. However, APS is an autoimmune disorder, and several approaches focused on modulating the immune response or its effectors have been employed. Those which have been most extensively studied include hydroxychloroquine, rituximab and eculizumab, an inhibitor of complement C5. In this report, we review in depth, and critique, key clinical studies of these agents. Since all of these studies are small, our conclusions are qualified. However, it appears that hydroxychloroquine may enhance the anticoagulant efficacy of vitamin K antagonists in APS patients, and that rituximab may ameliorate some of the "non-criteria" manifestations of APS. The catastrophic antiphospholipid syndrome (CAPS) is associated with diffuse thrombosis, multi-organ dysfunction, and ∼30% mortality. A high incidence of complement regulatory gene mutations, and compelling data concerning the efficacy of eculizumab in CAPS, suggests an important role for complement in this disorder. However, additional work is needed to clarify the role of complement in non-catastrophic APS, though emerging data suggests that complement inhibition may be effective in preventing thrombosis in these patients as well., (Copyright © 2022. Published by Elsevier Inc.)
- Published
- 2022
- Full Text
- View/download PDF
29. Race, rituximab, and relapse in TTP.
- Author
-
Chaturvedi S, Antun AG, Farland AM, Woods R, Metjian A, Park YA, de Ridder G, Gibson B, Kasthuri RS, Liles DK, Akwaa F, Clover T, Baumann Kreuziger L, Sadler JE, Sridharan M, Go RS, McCrae KR, Upreti HV, Liu A, Lim MY, Gangaraju R, Zheng XL, Raval JS, Masias C, Cataland SR, Johnson A, Davis E, Evans MD, and Mazepa MA
- Subjects
- ADAMTS13 Protein, Adrenal Cortex Hormones, Humans, Recurrence, Rituximab therapeutic use, Purpura, Thrombotic Thrombocytopenic therapy
- Abstract
Immune-mediated thrombotic thrombocytopenic purpura (iTTP) is characterized by recurring episodes of thrombotic microangiopathy, causing ischemic organ impairment. Black patients are overrepresented in iTTP cohorts in the United States, but racial disparities in iTTP outcome and response to therapy have not been studied. Using the United States Thrombotic Microangiopathies Consortium iTTP Registry, we evaluated the impact of race on mortality and relapse-free survival (RFS) in confirmed iTTP in the United States from 1995 to 2020. We separately examined the impact of rituximab therapy and presentation with newly diagnosed (de novo) or relapsed iTTP on RFS by race. A total of 645 participants with 1308 iTTP episodes were available for analysis. Acute iTTP mortality did not differ by race. When all episodes of iTTP were included, Black race was associated with shorter RFS (hazard ratio [HR], 1.60; 95% CI, 1.16-2.21); the addition of rituximab to corticosteroids improved RFS in White (HR, 0.37; 95% CI, 0.18-0.73) but not Black patients (HR, 0.96; 95% CI, 0.71-1.31). In de novo iTTP, rituximab delayed relapse, but Black patients had shorter RFS than White patients, regardless of treatment. In relapsed iTTP, rituximab significantly improved RFS in White but not Black patients. Race affects overall relapse risk and response to rituximab in iTTP. Black patients may require closer monitoring, earlier retreatment, and alternative immunosuppression after rituximab treatment. How race, racism, and social determinants of health contribute to the disparity in relapse risk in iTTP deserves further study., (© 2022 by The American Society of Hematology.)
- Published
- 2022
- Full Text
- View/download PDF
30. Development and performance of a hereditary hemorrhagic telangiectasia-specific quality-of-life instrument.
- Author
-
Kasthuri RS, Chaturvedi S, Thomas S, Vandergrift N, Bann C, Schaefer N, Clancy MS, Pyeritz R, and McCrae KR
- Subjects
- Cross-Sectional Studies, Epistaxis psychology, Humans, Quality of Life, Surveys and Questionnaires, Telangiectasia, Hereditary Hemorrhagic psychology
- Abstract
Hereditary hemorrhagic telangiectasia (HHT) is characterized by arteriovenous malformations and telangiectasia, with primary clinical manifestations of epistaxis and gastrointestinal bleeding and resultant anemia. HHT negatively affects health-related quality of life (HR-QoL); however, existing tools to measure HR-QoL are not HHT specific. Our objective was to develop an HHT-specific HR-QoL (HHT-QoL) instrument and evaluate its performance in a cross-sectional survey of individuals with HHT. Four HHT-specific questions were developed to evaluate the impact of HHT on productivity and social and personal interactions. An anonymous e-mail survey was conducted through Cure HHT. Participants also indicated their perceived HHT severity and completed 3 Patient-Reported Outcomes Measurement Information System (PROMIS) questionnaires: Discretionary Social Activities, Social Roles, and Emotional Distress. Complete data were available for 290 participants who self-identified their HHT severity as mild (29%), moderate (46%), or severe (25%). The HHT-QoL scale was reliable (Cronbach's-α, 0.83). Principal components analysis indicated the instrument was unidimensional. Participants had low levels of QoL with their ability to participate in discretionary social activities (PROMIS mean 36.4 [standard deviation 14.3]) and perform in social roles (41.5 [17.2]), and the presence of a high level of emotional distress (64.8 [24.2]). The HHT-QoL score correlated negatively with PROMIS Discretionary Social Activities (r = -0.65) and Social Roles (r = -0.68) and positively correlated with PROMIS Emotional Distress (r = 0.51). In conclusion, the 4-item HHT-QoL instrument provides valuable insight and may be a useful addition to future clinical research in HHT., (© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
31. Old is new again: emergence of thromboembolic complications in cancer patients on immunotherapy.
- Author
-
McCrae KR, Swaidani S, Diaz-Montero CM, and Khorana AA
- Subjects
- Humans, Immunologic Factors therapeutic use, Immunotherapy adverse effects, Inflammation drug therapy, Antineoplastic Agents, Immunological adverse effects, Neoplasms complications, Neoplasms drug therapy, Thrombosis drug therapy
- Abstract
Cancer immunotherapy has emerged as one of the most important new treatments for cancer in many years, moving rapidly to front-line therapy for several cancers. Cancer immunotherapy is based on treatment with immune checkpoint inhibitors (ICI), which are monoclonal antibodies directed toward immunoregulatory proteins including PD-1, PD-L1 and CTLA-4. ICI inhibit interactions between these proteins and their ligands, disabling physiologic immune regulatory networks and enhancing anti-tumor immunity. However, since the immune response cannot be directed specifically to the tumor, ICI are associated with immune-related adverse events (irAEs) resulting from immune-mediated attack of normal tissues. We and others have reported a high incidence of thrombosis in patients treated with ICI, which may approach 20%. Given the rapidly increasing use of ICIs, it is clear that I CI- A ssociated T hrombosis (IAT) is a major emerging clinical problem. However, there is a remarkable knowledge gap concerning mechanisms of IAT. IAT may be a composite irAE resulting from activation of blood and vascular cells, leading to thromboinflammation. Cancer itself is an inflammatory disorder, and inducing further inflammation through ICI administration may stimulate procoagulant activity by multiple cell types. Moreover, some blood and vascular cells express ICI target proteins. Here, we review the results of several studies describing the clinical manifestations of IAT, as well as our recent studies demonstrating that elevated levels of myeloid derived suppressor cells and inflammatory cytokines may serve as biomarkers of IAT. It is hoped that the concepts reviewed here may stimulate further research into this important clinical problem.
- Published
- 2022
- Full Text
- View/download PDF
32. Sclerotherapy Versus Cautery/Laser Treatment for Epistaxis in Hereditary Hemorrhagic Telangiectasia.
- Author
-
Woodard TD, Yappel-Sinkko KB, Wang X, McCrae KR, and Parambil JG
- Subjects
- Electrocoagulation adverse effects, Hemoglobins, Humans, Iron, Lasers, Postoperative Complications, Quality of Life, Retrospective Studies, Sclerotherapy methods, Epistaxis etiology, Epistaxis surgery, Telangiectasia, Hereditary Hemorrhagic therapy
- Abstract
Objectives/hypothesis: Surgical interventions for epistaxis management in hereditary hemorrhagic telangiectasia (HHT) demonstrate short-term success and require repeated procedures for disease control. Although electrocautery and/or laser photocoagulation (C ± L) are most frequently performed, sodium tetradecyl sclerotherapy (STS) is emerging as a promising newer treatment. We hypothesized that in a 24-month time period, STS would require fewer treatments than C ± L to maintain epistaxis severity within the mild range., Study Design: Retrospective study., Methods: We retrospectively assessed 67 patients with HHT with moderate and severe epistaxis that were treated periodically with C ± L (34 patients) versus STS (33 patients). The primary outcome was the number of procedures needed to maintain the epistaxis severity score (ESS) as mild. Secondary outcomes assessed for differences in postoperative complications, hemoglobin levels, iron stores, hematologic support, and quality-of-life (QoL) scores., Results: To maintain ESS in the mild range, 1.6 STS procedures (range, 1-4) were performed versus 3.6 C ± L procedures (range, 1-8) (P = .003). Significant postoperative differences included reduction in nasal crusting (3% vs. 32%, P = .001), foul odor (3% vs. 35%, P < .001), and septal perforation (3% vs. 29%, P = .006) after STS. There were no significant differences between the two treatments in hemoglobin levels, iron stores, hematologic support, or QoL scores., Conclusion: STS is able to attain satisfactory epistaxis control with significantly fewer procedures and lower postoperative complications than C ± L. STS should be considered as the initial surgical intervention for epistaxis in patients with HHT., Level of Evidence: 4 Laryngoscope, 132:920-925, 2022., (© 2021 The American Laryngological, Rhinological and Otological Society, Inc.)
- Published
- 2022
- Full Text
- View/download PDF
33. Eculizumab for refractory thrombosis in antiphospholipid syndrome.
- Author
-
Hussain H, Tarantino MD, Chaturvedi S, McCrae KR, and Roberts JC
- Subjects
- Adolescent, Antibodies, Monoclonal, Humanized, Aspirin therapeutic use, Clopidogrel therapeutic use, Female, Fondaparinux therapeutic use, Humans, Hydroxychloroquine therapeutic use, Antiphospholipid Syndrome complications, Antiphospholipid Syndrome drug therapy, Thrombosis drug therapy, Thrombosis etiology
- Abstract
Antiphospholipid syndrome (APS) is characterized by arterial and/or venous thrombosis with antiphospholipid antibodies. Dysregulation of the complement pathway has been implicated in APS pathophysiology. We report the successful use of eculizumab, an anti-C5 monoclonal antibody, in controlling and preventing recurrent thrombosis in a refractory case of APS. An 18-year-old female was diagnosed with APS after developing extensive, unprovoked deep vein thrombosis (DVT) of axillary, inferior vena cava, and brachiocephalic veins. Thrombophilia evaluation revealed triple-positive lupus anticoagulant, β-2 glycoprotein IgM, IgA, and anticardiolipin antibodies (each >40 U/mL) with persistently positive titers after 12 weeks. She was refractory to multiple anticoagulants alone (enoxaparin, fondaparinux, apixaban, rivaroxaban, and warfarin) with antiplatelet (aspirin and clopidogrel) and adjunctive therapies (hydroxychloroquine, immunosuppression with steroids and rituximab, and plasmapheresis). Despite these, she continued to develop recurrent thrombosis and additionally developed hepatic infarction and pulmonary embolism with failure to decrease titers after 6 weeks of plasma exchange. Following this event, eculizumab (600 mg weekly × 4 weeks followed by 900 mg every 2 weeks) was initiated in combination with fondaparinux, aspirin, clopidogrel, and hydroxychloroquine. She has remained on this regimen without recurrence of thrombosis. Our case suggests that eculizumab may have a role as a therapeutic option in refractory thrombosis in APS., (© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
34. Biomarker signatures in cancer patients with and without venous thromboembolism events: a substudy of CASSINI.
- Author
-
Khorana AA, Barnard J, Wun T, Vijapurkar U, Damaraju CV, Moore KT, Wildgoose P, and McCrae KR
- Subjects
- Bayes Theorem, Biomarkers, Female, Humans, Male, Prospective Studies, Thyrotropin, Neoplasms complications, Venous Thromboembolism diagnosis, Venous Thromboembolism etiology
- Abstract
Cancer is associated with an increased risk of venous thromboembolism (VTE). In the CASSINI study, ambulatory cancer patients with a Khorana risk score ≥2 had a reduced risk of VTE while receiving rivaroxaban. This analysis used blood samples from CASSINI to compare biomarker levels between patients with and without VTE. VTE occurred in 62 patients during the 6 months of CASSINI (cases), and they were matched by age, sex, cancer type, tumor stage, and Khorana score to 62 controls. Baseline blood samples were analyzed for 280 biomarkers, and biomarker distribution was compared using the Wilcoxon rank-sum test between groups defined by VTE occurrence and vital status. Sparse Bayesian regression modeling was used to select a joint panel of potential VTE biomarkers. Biomarkers with the largest differences in baseline distribution among cancer patients with and without VTE included decreases in stromal cell-derived factor-1 (SDF-1), thyroid-stimulating hormone (TSH), and monocyte chemotactic protein 4 and increases in growth hormone (GH) and interleukin-1 receptor type 1 (IL-1R1). Between survivors and those who died, significantly different biomarkers included ST2, IL-8, and C-reactive protein. Regression analyses also identified decreases in SDF-1 and TSH. Pathway analysis indicated enrichment of cytokine and chemokine activity with IL-1R1, SDF-1, and GH, which are the strongest predictors of VTE or death. Our analyses highlight the interactions between hemostatic and inflammatory processes and identify candidate biomarkers of cancer-associated VTE. Prospective studies will determine clinical relevance of these biomarkers. This trial was registered at www.ClinicalTrials.gov as #NCT02555878., (© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
35. Pazopanib for severe bleeding and transfusion-dependent anemia in hereditary hemorrhagic telangiectasia.
- Author
-
Parambil JG, Gossage JR, McCrae KR, Woodard TD, Menon KVN, Timmerman KL, Pederson DP, Sprecher DL, and Al-Samkari H
- Subjects
- Epistaxis drug therapy, Epistaxis etiology, Humans, Indazoles, Pyrimidines, Retrospective Studies, Sulfonamides, Anemia drug therapy, Anemia etiology, Telangiectasia, Hereditary Hemorrhagic complications, Telangiectasia, Hereditary Hemorrhagic drug therapy
- Abstract
Hereditary hemorrhagic telangiectasia (HHT) is a rare angiogenic disorder causing chronic gastrointestinal bleeding, epistaxis, and severe anemia. Pazopanib is an oral multi-kinase angiogenesis inhibitor with promise to treat bleeding in HHT. We analyzed outcomes of HHT patients with the most severe bleeding causing RBC transfusion dependence treated on a predefined institutional pazopanib treatment pathway (with data collected retrospectively). The primary endpoint was achievement of transfusion independence. Secondary endpoints included hemoglobin, epistaxis severity score, RBC transfusion and iron infusion requirements, number of local hemostatic procedures, ferritin and transferrin saturation, compared using paired and repeated measures mean tests. Thirteen transfusion-dependent HHT patients received pazopanib [median (range) dose 150 (25-300) mg daily)] for a median of 22 months. All patients achieved transfusion independence. Compared with pretreatment, pazopanib increased mean hemoglobin by 4.8 (95% CI, 3.6-5.9) g/dL (7.8 vs. 12.7 g/dL, P < 0.0001) and decreased mean epistaxis severity score by 4.77 (3.11-6.44) points (7.20 vs. 2.43 points, P < 0.0001) after 12 months of treatment. Compared with 3 months of pretreatment, RBC transfusions decreased by 93% (median of 16.0 vs. 0.0 units, P < 0.0001) and elemental iron infusion decreased by 92% (median of 4500 vs. 0 mg, P = 0.005) during the first 3 months of treatment; improvements were maintained over time. Pazopanib was well-tolerated: hypertension, lymphocytopenia, and fatigue were the most common TEAEs. In conclusion, pazopanib was safe and effective to manage severe bleeding in HHT, liberating all patients from transfusion dependence and normalizing hematologic parameters at doses lower than used to treat malignancies. These findings require confirmation in a randomized trial., (© 2021. The Author(s), under exclusive licence to Springer Nature B.V.)
- Published
- 2022
- Full Text
- View/download PDF
36. Effect of aspirin on short-term outcomes in hospitalized patients with COVID-19.
- Author
-
Sahai A, Bhandari R, Godwin M, McIntyre T, Chung MK, Iskandar JP, Kamran H, Hariri E, Aggarwal A, Burton R, Kalra A, Bartholomew JR, McCrae KR, Elbadawi A, Bena J, Svensson LG, Kapadia S, and Cameron SJ
- Subjects
- Adult, Aged, Anti-Inflammatory Agents, Non-Steroidal adverse effects, Aspirin adverse effects, Female, Hospitalization, Humans, Male, Middle Aged, Pandemics, SARS-CoV-2, Thrombosis virology, Anti-Inflammatory Agents, Non-Steroidal administration & dosage, Aspirin administration & dosage, COVID-19 complications, Inpatients, Thrombosis prevention & control
- Abstract
Coronavirus disease 2019 (COVID-19) caused by SARS-CoV-2 is an ongoing viral pandemic marked by increased risk of thrombotic events. However, the role of platelets in the elevated observed thrombotic risk in COVID-19 and utility of antiplatelet agents in attenuating thrombosis is unknown. We aimed to determine if the antiplatelet effect of aspirin may mitigate risk of myocardial infarction, cerebrovascular accident, and venous thromboembolism in COVID-19. We evaluated 22,072 symptomatic patients tested for COVID-19. Propensity-matched analyses were performed to determine if treatment with aspirin or nonsteroidal anti-inflammatory drugs (NSAIDs) affected thrombotic outcomes in COVID-19. Neither aspirin nor NSAIDs affected mortality in COVID-19. Thus, aspirin does not appear to prevent thrombosis and death in COVID-19. The mechanisms of thrombosis in COVID-19, therefore, appear distinct and the role of platelets as direct mediators of SARS-CoV-2-mediated thrombosis warrants further investigation.
- Published
- 2021
- Full Text
- View/download PDF
37. Polyphosphate expression by cancer cell extracellular vesicles mediates binding of factor XII and contact activation.
- Author
-
Shim YJ, Chatterjee V, Swaidani S, Alluri RK, Kundu S, Merkulova A, Angelini D, You D, Whitney SA, Feener EP, Barnard J, Schmaier AH, Khorana AA, and McCrae KR
- Subjects
- Animals, Factor XII, Factor XIIa, Humans, Mice, Polyphosphates, Prekallikrein, Extracellular Vesicles, Neoplasms
- Abstract
Extracellular vesicles (EV) have been implicated in diverse biological processes, including intracellular communication, transport of nucleic acids, and regulation of vascular function. Levels of EVs are elevated in cancer, and studies suggest that EV may stimulate thrombosis in patients with cancer through expression of tissue factor. However, limited data also implicate EV in the activation of the contact pathway of coagulation through activation of factor XII (FXII) to FXIIa. To better define the ability of EV to initiate contact activation, we compared the ability of EV derived from different cancer cell lines to activate FXII. EV from all cell lines activated FXII, with those derived from pancreatic and lung cancer cell lines demonstrating the most potent activity. Concordant with the activation of FXII, EV induced the cleavage of high molecular weight kininogen (HK) to cleaved kininogen. We also observed that EVs from patients with cancer stimulated FXII activation and HK cleavage. To define the mechanisms of FXII activation by EV, EV were treated with calf intestinal alkaline phosphatase or Escherichia coli exopolyphosphatase to degrade polyphosphate; this treatment blocked binding of FXII to EVs and the ability of EV to mediate FXII activation. In vivo, EV induced pulmonary thrombosis in wild-type mice, with protection conferred by a deficiency in FXII, HK, or prekallikrein. Moreover, pretreatment of EVs with calf intestinal alkaline phosphatase inhibited their prothrombotic effect. These results indicate that polyphosphate mediates the binding of contact factors to EV and that EV-associated polyphosphate may contribute to the prothrombotic effects of EV in cancer., (© 2021 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
38. Plasmin-mediated cleavage of high-molecular-weight kininogen contributes to acetaminophen-induced acute liver failure.
- Author
-
Henderson MW, Sparkenbaugh EM, Wang S, Ilich A, Noubouossie DF, Mailer R, Renné T, Flick MJ, Luyendyk JP, Chen ZL, Strickland S, Stravitz RT, McCrae KR, Key NS, and Pawlinski R
- Subjects
- Acetaminophen pharmacology, Animals, Chemical and Drug Induced Liver Injury genetics, Chemical and Drug Induced Liver Injury pathology, Factor XII genetics, Factor XII metabolism, Female, Fibrinolysin genetics, Humans, Kininogens genetics, Male, Mice, Mice, Knockout, Prekallikrein genetics, Prekallikrein metabolism, Acetaminophen adverse effects, Chemical and Drug Induced Liver Injury metabolism, Fibrinolysin metabolism, Fibrinolysis drug effects, Kininogens metabolism, Proteolysis drug effects
- Abstract
Acetaminophen (APAP)-induced liver injury is associated with activation of coagulation and fibrinolysis. In mice, both tissue factor-dependent thrombin generation and plasmin activity have been shown to promote liver injury after APAP overdose. However, the contribution of the contact and intrinsic coagulation pathways has not been investigated in this model. Mice deficient in individual factors of the contact (factor XII [FXII] and prekallikrein) or intrinsic coagulation (FXI) pathway were administered a hepatotoxic dose of 400 mg/kg of APAP. Neither FXII, FXI, nor prekallikrein deficiency mitigated coagulation activation or hepatocellular injury. Interestingly, despite the lack of significant changes to APAP-induced coagulation activation, markers of liver injury and inflammation were significantly reduced in APAP-challenged high-molecular-weight kininogen-deficient (HK-/-) mice. Protective effects of HK deficiency were not reproduced by inhibition of bradykinin-mediated signaling, whereas reconstitution of circulating levels of HK in HK-/- mice restored hepatotoxicity. Fibrinolysis activation was observed in mice after APAP administration. Western blotting, enzyme-linked immunosorbent assay, and mass spectrometry analysis showed that plasmin efficiently cleaves HK into multiple fragments in buffer or plasma. Importantly, plasminogen deficiency attenuated APAP-induced liver injury and prevented HK cleavage in the injured liver. Finally, enhanced plasmin generation and HK cleavage, in the absence of contact pathway activation, were observed in plasma of patients with acute liver failure due to APAP overdose. In summary, extrinsic but not intrinsic pathway activation drives the thromboinflammatory pathology associated with APAP-induced liver injury in mice. Furthermore, plasmin-mediated cleavage of HK contributes to hepatotoxicity in APAP-challenged mice independently of thrombin generation or bradykinin signaling., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
39. Stress Granule-Mediated Oxidized RNA Decay in P-Body: Hypothetical Role of ADAR1, Tudor-SN, and STAU1.
- Author
-
Alluri RK, Li Z, and McCrae KR
- Abstract
Reactive oxygen species (ROS) generated under oxidative stress (OS) cause oxidative damage to RNA. Recent studies have suggested a role for oxidized RNA in several human disorders. Under the conditions of oxidative stress, mRNAs released from polysome dissociation accumulate and initiate stress granule (SG) assembly. SGs are highly enriched in mRNAs, containing inverted repeat (IR) Alus in 3' UTRs, AU-rich elements, and RNA-binding proteins. SGs and processing bodies (P-bodies) transiently interact through a docking mechanism to allow the exchange of RNA species. However, the types of RNA species exchanged, and the mechanisms and outcomes of exchange are still unknown. Specialized RNA-binding proteins, including adenosine deaminase acting on RNA (ADAR1-p150), with an affinity toward inverted repeat Alus , and Tudor staphylococcal nuclease (Tudor-SN) are specifically recruited to SGs under OS along with an RNA transport protein, Staufen1 (STAU1), but their precise biochemical roles in SGs and SG/P-body docking are uncertain. Here, we critically review relevant literature and propose a hypothetical mechanism for the processing and decay of oxidized-RNA in SGs/P-bodies, as well as the role of ADAR1-p150, Tudor-SN, and STAU1., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Alluri, Li and McCrae.)
- Published
- 2021
- Full Text
- View/download PDF
40. Efficacy and Safety of Avatrombopag in Patients with Chronic Immune Thrombocytopenia: A Systematic Literature Review and Network Meta-Analysis.
- Author
-
Wojciechowski P, Wilson K, Nazir J, Pustułka I, Tytuła A, Smela B, Pochopien M, Vredenburg M, McCrae KR, and Jurczak W
- Subjects
- Benzoates, Humans, Network Meta-Analysis, Randomized Controlled Trials as Topic, Recombinant Fusion Proteins, Thiazoles, Thiophenes, Purpura, Thrombocytopenic, Idiopathic drug therapy
- Abstract
Introduction: A network meta-analysis (NMA) was performed to assess the efficacy and safety of avatrombopag, relative to eltrombopag, romiplostim, and fostamatinib, for patients with chronic immune thrombocytopenia (ITP) not responding adequately to corticosteroids., Methods: A systematic search of publication and clinical trial databases was conducted to identify relevant randomized controlled trials (RCTs) and observational studies. Data from eligible studies were extracted and analyzed in a Bayesian framework using relative effect sizes vs placebo. Outcomes included durable platelet response; need for rescue therapy; reduction in use of concomitant ITP medication; incidence of any or World Health Organization (WHO) grade 2-4 bleeding events, and any adverse events. Results were reported as odds ratios or incidence rate ratios (IRR) with 95% credible intervals (CrIs)., Results: The NMA included seven phase 3 RCTs. Compared with placebo, avatrombopag was associated with statistically significant improvements in durable platelet response, reduction in use of concomitant ITP medication, and incidence of any bleeding events. Statistically significant differences vs placebo were also observed for durable platelet response and need for rescue therapy (eltrombopag, romiplostim, and fostamatinib); reduction in use of concomitant ITP medication (eltrombopag and romiplostim); incidence of any bleeding events (fostamatinib); and incidence of WHO grade 2-4 bleeding events (romiplostim and fostamatinib). No statistically significant differences were observed for any adverse events. Avatrombopag was associated with a statistically significant lower incidence of any bleeding events vs eltrombopag (IRR 0.38 [95% CrI 0.19, 0.75]) and romiplostim (IRR 0.38 [95% Crl 0.17, 0.86]); no other between-treatment differences were observed., Conclusion: In this NMA, avatrombopag significantly increased the chance of achieving durable platelet response and reducing the use of concomitant ITP medication vs placebo, and significantly reduced the incidence of any bleeding events compared with placebo, eltrombopag, and romiplostim. The study aims to help guide clinicians managing patients with chronic ITP and insufficient response to previous treatment.
- Published
- 2021
- Full Text
- View/download PDF
41. Thrombotic thrombocytopenia due to SARS-CoV-2 vaccination.
- Author
-
McCrae KR
- Abstract
Vaccine-induced thrombotic thrombocytopenia (VITT) has been reported after vaccination with the AztraZeneca ChAdOx1 nCoV-19 and the Johnson and Johnson Ad26. COV2.S vaccines. This manuscript provides a brief overview of reported cases, clinical and laboratory features, and current understanding of the pathogenesis of VITT. The author also poses unananswered questions and identifies directions for future study., (Copyright © 2021 The Cleveland Clinic Foundation. All Rights Reserved.)
- Published
- 2021
- Full Text
- View/download PDF
42. A novel "vascular" thrombophilia.
- Author
-
Alluri RK and McCrae KR
- Subjects
- Humans, Risk Factors, Thrombophilia
- Published
- 2021
- Full Text
- View/download PDF
43. Increased Incidence of Venous Thromboembolism with Cancer Immunotherapy.
- Author
-
Roopkumar J, Swaidani S, Kim AS, Thapa B, Gervaso L, Hobbs BP, Wei W, Alban TJ, Funchain P, Kundu S, Sangwan N, Rayman P, Pavicic PG Jr, Diaz-Montero CM, Barnard J, McCrae KR, and Khorana AA
- Subjects
- Humans, Immunotherapy adverse effects, Incidence, Interleukin-8 therapeutic use, Retrospective Studies, Neoplasms complications, Venous Thromboembolism epidemiology
- Abstract
Background: Cancer immunotherapy is associated with several immune-related adverse events, but the relationship between immunotherapy and venous thromboembolism has not been thoroughly studied., Methods: We conducted a retrospective cohort study of 1,686 patients who received immunotherapy for a variety of malignancies to determine the incidence of venous thromboembolism and the impact of venous thromboembolism on survival. To examine the potential role of inflammation in venous thromboembolism, we also profiled immune cells and plasma cytokines in blood samples obtained prior to initiation of immunotherapy in a sub-cohort of patients treated on clinical trials who subsequently did (N = 15), or did not (N = 10) develop venous thromboembolism., Findings: Venous thromboembolism occurred while on immunotherapy in 404/1686 patients (24%) and was associated with decreased overall survival [HR=1.22 (95% CI 1.06-1.41), p<0.008]. Patients that developed venous thromboembolism had significantly higher pretreatment levels of myeloid-derived suppressor cells (5.382 ± 0.873 vs. 3.341 ± 0.3402, mean ± SEM; p=0.0045), interleukin 8 (221.2 ± 37.53 vs. 111.6 ± 25.36, mean ± SEM; p=0.016), and soluble vascular cell adhesion protein 1 (1210 ± 120.6 vs. 895.5 ± 53.34, mean ± SEM; p=0.0385)., Conclusions: These findings demonstrate that venous thromboembolism is an underappreciated and important immune-related adverse event associated with cancer immunotherapy, and may implicate an interleukin 8 and myeloid-derived suppressor cell-driven pathway in pathogenesis.
- Published
- 2021
- Full Text
- View/download PDF
44. Risk of thromboembolism in patients with ALK- and EGFR-mutant lung cancer: A cohort study.
- Author
-
Roopkumar J, Poudel SK, Gervaso L, Reddy CA, Velcheti V, Pennell NA, McCrae KR, and Khorana AA
- Subjects
- Anaplastic Lymphoma Kinase genetics, Anticoagulants, Cohort Studies, ErbB Receptors genetics, Female, Humans, Lymphocytes, Male, Mutation, Receptor Protein-Tyrosine Kinases, Retrospective Studies, Carcinoma, Non-Small-Cell Lung genetics, Lung Neoplasms genetics, Venous Thromboembolism diagnosis, Venous Thromboembolism epidemiology, Venous Thromboembolism genetics
- Abstract
Introduction: Thromboembolism (TE) is common in patients with non-small cell lung cancer (NSCLC) and is associated with worse outcomes. Recent advances in the understanding of NSCLC have led to the identification of molecular subtypes such as anaplastic lymphocyte kinase (ALK) and epidermal growth factor receptor (EGFR) mutations. The association of these subtypes with risk of TE has not been fully explored., Methods: We conducted a retrospective cohort study of consecutive NSCLC patients seen at the Cleveland Clinic from July 2002 through July 2017 for whom molecular classification and follow-up were available. TE events included deep vein thrombosis (DVT), pulmonary embolism (PE), visceral vein thrombosis (VVT), and arterial events. TE-free survival and overall survival rates for each of the molecular subtypes (wild-type, ALK-mutant, and EGFR-mutant) were estimated by the Kaplan-Meier method. Cox proportional hazard regression analysis was used to identify factors associated with the endpoints TE and overall survival. TE was analyzed as a conditional, time-dependent covariate to assess its impact with respect to overall survival., Results: The study population consisted of 461 patients. Approximately half were females (n = 263, 57%) and 58% (n = 270) were older than 65 years. TE occurred in 98 of 461 patients (21.3%) during a median follow-up of 33.1 months. The highest cumulative rates of TE were observed in patients with ALK-mutant NSCLC (N = 20/46, 43.5%) followed by patients with EGFR-mutant cancers (N = 35/165, 21.2%) and wild-type cancers (N = 43/250, 17.2%) P < .05. Cumulative incidence of TE at 6 months of follow-up was 15.7% (95% confidence interval [CI]: 5.0%-26.4%) for ALK-mutant cancers, 8.8% (95% CI: 4.4%-13.2%) for EGFR-mutant cancers, and 9.2% (95% CI: 5.4%-12.9%) for wild-type cancers. Patients who experienced TE had worse overall survival (all patients: hazard ratio = 2.8 95% CI 2.1-3.6, P < .001)., Conclusions: Patients with ALK-mutant advanced lung adenocarcinoma have the highest rate of TE. TE is associated with worse survival across molecular subtypes. These findings should be taken into consideration in decision-making regarding thromboprophylaxis., (© 2020 International Society on Thrombosis and Haemostasis.)
- Published
- 2021
- Full Text
- View/download PDF
45. Incidence of thromboembolism in patients with melanoma on immune checkpoint inhibitor therapy and its adverse association with survival.
- Author
-
Sussman TA, Li H, Hobbs B, Funchain P, McCrae KR, and Khorana AA
- Subjects
- Adult, Aged, Aged, 80 and over, Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, Female, Humans, Immune Checkpoint Inhibitors adverse effects, Incidence, Ipilimumab administration & dosage, Ipilimumab adverse effects, Ischemic Attack, Transient chemically induced, Male, Melanoma pathology, Middle Aged, Nivolumab administration & dosage, Nivolumab adverse effects, Retrospective Studies, Risk Factors, Survival Analysis, Thromboembolism chemically induced, Treatment Outcome, Young Adult, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Immune Checkpoint Inhibitors administration & dosage, Ischemic Attack, Transient epidemiology, Melanoma drug therapy, Thromboembolism epidemiology
- Abstract
Background: Thromboembolism (TE) in cancer significantly contributes to morbidity and mortality. Little is known about the incidence of arterial TE (ATE) and venous TE (VTE) in patients with melanoma on immune checkpoint inhibitor (ICI) therapy., Methods: We conducted a retrospective cohort study of patients with melanoma receiving ICI from July 2015 through December 2017 at the Cleveland Clinic. TE, including VTE events of deep venous thrombosis, pulmonary embolism, visceral vein thrombosis, and ATE events of myocardial infarction, stroke, peripheral arterial embolism, or transient ischemic attack after ICI initiation were identified. Overall survival (OS) from ICI initiation was estimated by Kaplan-Meier and Cox hazard models; associations between TE, ICI regimen, and clinical risk factors were evaluated using log-rank test., Results: The study population comprised 228 patients with median age of 65 years (23-91 years), 67% male, and median follow-up of 27.3 months. Pembrolizumab was most commonly used (38.7%), followed by combination of ipilimumab plus nivolumab (29.4%), ipilimumab (20%), and nivolumab (12.3%). Most had stage IV disease (81.1%) and 11% had brain metastases (BM) at treatment initiation. Fifty-one TE events occurred in 47 patients (20.6%), including 37 (16.2%) VTE and 14 (6.1%) ATE. Cumulative incidence of TE after ICI initiation was 9.3% (95% CI: 6.0% to 13.6%) at 6 months, and 16.0% (95% CI: 11.6% to 21.2%) at 12 months. The 6-month and 12-month VTE cumulative incidence rates were higher with combination ICI than single agent (16.7% vs 5.0% and 21.3% vs 9.5%, respectively; p=0.02). Risk factors significantly associated with VTE in multivariate analysis included combination ICI (HR 2.70; 95% CI: 1.28 to 5.70; p=0.009), Khorana Score ≥1 (HR 2.24; 95% CI: 1.06 to 4.74; p=0.03), history of coronary artery disease (HR 2.71; 95% CI: 1.16 to 6.29); p=0.02), and anticoagulation at treatment start (HR 4.14; 95% CI: 1.60 to 10.7; p=0.003). Of patients without BM, OS was worse in patients with TE compared with those without (2-year OS 50.8% vs 71.3%; HR 2.27; 95% CI: 1.36 to 3.79; p=0.002), when adjusted for age and stage., Conclusions: ICI is associated with a high incidence of TE in patients with melanoma, with higher rates with combination therapy; TE is associated with substantial worsening of survival. Further studies are needed to identify pathophysiology, biomarkers, and preventive approaches., Competing Interests: Competing interests: PF receives institutional research funding from Pfizer and Bristol Myers Squibb, outside the submitted work. BH reports research funds from Scientific Advisory for Bayer AG, Presagia, and STCube Pharmaceuticals, outside the submitted work. AAK reports personal fees and non-financial support from Janssen, Bayer, Sanofi, Halozyme, Seattle Genetics, Pfizer, and Medscape, personal fees from Parexel and TriSalus, grants from Merck, Array, and Leap, and grants and personal fees from Bristol Myers Squibb, outside the submitted work., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2021
- Full Text
- View/download PDF
46. SARS-CoV-2 Receptors are Expressed on Human Platelets and the Effect of Aspirin on Clinical Outcomes in COVID-19 Patients.
- Author
-
Sahai A, Bhandari R, Koupenova M, Freedman JE, Godwin M, McIntyre T, Chung MK, Iskandar JP, Kamran H, Hariri E, Aggarwal A, Kalra A, Bartholomew JR, McCrae KR, Elbadawi A, Svensson LG, Kapadia S, and Cameron SJ
- Abstract
Coronavirus disease-2019 (COVID-19) caused by SARS-CoV-2 is an ongoing viral pandemic marked by increased risk of thrombotic events. However, the role of platelets in the elevated observed thrombotic risk in COVID-19 and utility of anti-platelet agents in attenuating thrombosis is unknown. We aimed to determine if human platelets express the known SARS-CoV-2 receptor-protease axis on their cell surface and assess whether the anti-platelet effect of aspirin may mitigate risk of myocardial infarction (MI), cerebrovascular accident (CVA), and venous thromboembolism (VTE) in COVID-19. Expression of ACE2 and TMPRSS2 on human platelets were detected by immunoblotting and confirmed by confocal microscopy. We evaluated 22,072 symptomatic patients tested for COVID-19. Propensity-matched analyses were performed to determine if treatment with aspirin or non-steroidal anti-inflammatory drugs (NSAIDs) affected thrombotic outcomes in COVID-19. Neither aspirin nor NSAIDs affected mortality in COVID-19. However, both aspirin and NSAID therapies were associated with increased risk of the combined thrombotic endpoint of (MI), (CVA), and (VTE). Thus, while platelets clearly express ACE2-TMPRSS2 receptor-protease axis for SARS-CoV-2 infection, aspirin does not prevent thrombosis and death in COVID-19. The mechanisms of thrombosis in COVID-19, therefore, appears distinct and the role of platelets as direct mediators of SARS-CoV-2-mediated thrombosis warrants further investigation., Competing Interests: Declaration of Interests None of the authors have any relevant conflicting financial, personal, or professional relationships.
- Published
- 2020
- Full Text
- View/download PDF
47. Tapering thrombopoietin receptor agonists in primary immune thrombocytopenia: Expert consensus based on the RAND/UCLA modified Delphi panel method.
- Author
-
Cuker A, Despotovic JM, Grace RF, Kruse C, Lambert MP, Liebman HA, Lyons RM, McCrae KR, Pullarkat V, Wasser JS, Beenhouwer D, Gibbs SN, Yermilov I, and Broder MS
- Abstract
Background: Thrombopoietin receptor agonists (TPO-RAs) are used to treat primary immune thrombocytopenia (ITP). Some patients have discontinued treatment while maintaining a hemostatic platelet count., Objectives: To develop expert consensus on when it is appropriate to consider tapering TPO-RAs in ITP, how to taper patients off therapy, how to monitor patients after discontinuation, and how to restart therapy., Methods: We used a RAND/UCLA modified Delphi panel method. Ratings were completed independently by each expert before and after a meeting. Second-round ratings were used to develop the panel's guidance. The panel was double-blinded: The sponsor and nonchair experts did not know each other's identities., Results: Guidance on when it is appropriate to taper TPO-RAs in children and adults was developed based on patient platelet count, history of bleeding, intensification of treatment, trauma risk, and use of anticoagulants/platelet inhibitors. For example, it is appropriate to taper TPO-RAs in patients who have normal/above-normal platelet counts, have no history of major bleeding, and have not required an intensification of treatment in the past 6 months; it is inappropriate to taper TPO-RAs in patients with low platelet counts. Duration of ITP, months on TPO-RA, or timing of platelet response to TPO-RA did not have an impact on the panel's guidance on appropriateness to taper. Guidance on how to taper patients off therapy, how to monitor patients after discontinuation, and how to restart therapy is also provided., Conclusion: This guidance could support clinical decision making and the development of clinical trials that prospectively test the safety of tapering TPO-RAs., (© 2020 The Authors. Research and Practice in Thrombosis and Haemostasis published by Wiley Periodicals LLC on behalf of International Society on Thrombosis and Haemostasis.)
- Published
- 2020
- Full Text
- View/download PDF
48. Mechanisms and biomarkers of cancer-associated thrombosis.
- Author
-
Kim AS, Khorana AA, and McCrae KR
- Subjects
- Anticoagulants therapeutic use, Humans, Neoplasms metabolism, Thrombosis drug therapy, Thrombosis metabolism, Biomarkers, Tumor metabolism, Neoplasms complications, Thrombosis complications
- Abstract
Cancer-associated thrombosis is a leading cause of non-cancer death in cancer patients and is comprised of both arterial and venous thromboembolism (VTE). There are multiple risk factors for developing VTE, including cancer type, stage, treatment, and other medical comorbidities, which suggests that the etiology of thrombosis is multifactorial. While cancer-associated thrombosis can be treated with anticoagulation, benefits of therapy must be balanced with the increased bleeding risks seen in patients with cancer. Although risk models exist for primary and recurrent VTE, additional predictors are needed to improve model performance and discrimination of high-risk patients. This review will outline the diverse mechanisms driving thrombosis in cancer patients, as well as provide an overview of biomarkers studied in thrombosis risk and important considerations when selecting candidate biomarkers., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
49. Factor XII and kininogen asymmetric assembly with gC1qR/C1QBP/P32 is governed by allostery.
- Author
-
Kaira BG, Slater A, McCrae KR, Dreveny I, Sumya U, Mutch NJ, Searle M, and Emsley J
- Subjects
- Aged, Carrier Proteins metabolism, Factor XII metabolism, Female, Humans, Kinetics, Kininogens metabolism, Ligands, Membrane Glycoproteins metabolism, Mitochondrial Proteins metabolism, Models, Biological, Molecular Dynamics Simulation, Multiprotein Complexes chemistry, Multiprotein Complexes metabolism, Protein Binding, Protein Conformation, Receptors, Complement metabolism, Recombinant Proteins, Structure-Activity Relationship, Zinc chemistry, Zinc metabolism, Allosteric Site, Binding Sites, Carrier Proteins chemistry, Factor XII chemistry, Kininogens chemistry, Membrane Glycoproteins chemistry, Mitochondrial Proteins chemistry, Models, Molecular, Receptors, Complement chemistry
- Abstract
The contact system is composed of factor XII (FXII), prekallikrein (PK), and cofactor high-molecular-weight kininogen (HK). The globular C1q receptor (gC1qR) has been shown to interact with FXII and HK. We reveal the FXII fibronectin type II domain (FnII) binds gC1qR in a Zn2+-dependent fashion and determined the complex crystal structure. FXIIFnII binds the gC1qR trimer in an asymmetric fashion, with residues Arg36 and Arg65 forming contacts with 2 distinct negatively charged pockets. gC1qR residues Asp185 and His187 coordinate a Zn2+ adjacent to the FXII-binding site, and a comparison with the ligand-free gC1qR crystal structure reveals the anionic G1-loop becomes ordered upon FXIIFnII binding. Additional conformational changes in the region of the Zn2+-binding site reveal an allosteric basis for Zn2+ modulation of FXII binding. Mutagenesis coupled with surface plasmon resonance demonstrate the gC1qR Zn2+ site contributes to FXII binding, and plasma-based assays reveal gC1qR stimulates coagulation in a FXII-dependent manner. Analysis of the binding of HK domain 5 (HKD5) to gC1qR shows only 1 high-affinity binding site per trimer. Mutagenesis studies identify a critical G3-loop located at the center of the gC1qR trimer, suggesting steric occlusion as the mechanism for HKD5 asymmetric binding. Gel filtration experiments reveal that gC1qR clusters FXII and HK into a higher-order 500-kDa ternary complex. These results support the conclusion that extracellular gC1qR can act as a chaperone to cluster contact factors, which may be a prelude for initiating the cascades that drive bradykinin generation and the intrinsic pathway of coagulation., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
50. SARS-CoV-2 and ACE2: The biology and clinical data settling the ARB and ACEI controversy.
- Author
-
Chung MK, Karnik S, Saef J, Bergmann C, Barnard J, Lederman MM, Tilton J, Cheng F, Harding CV, Young JB, Mehta N, Cameron SJ, McCrae KR, Schmaier AH, Smith JD, Kalra A, Gebreselassie SK, Thomas G, Hawkins ES, and Svensson LG
- Subjects
- Angiotensin Receptor Antagonists pharmacology, Angiotensin-Converting Enzyme 2, Angiotensin-Converting Enzyme Inhibitors pharmacology, Animals, Betacoronavirus pathogenicity, COVID-19, Coronavirus Infections metabolism, Coronavirus Infections pathology, Coronavirus Infections virology, Humans, Kallikrein-Kinin System drug effects, Pandemics, Peptidyl-Dipeptidase A genetics, Pneumonia, Viral metabolism, Pneumonia, Viral pathology, Pneumonia, Viral virology, Renin-Angiotensin System drug effects, SARS-CoV-2, Angiotensin Receptor Antagonists therapeutic use, Angiotensin-Converting Enzyme Inhibitors therapeutic use, Coronavirus Infections drug therapy, Peptidyl-Dipeptidase A metabolism, Pneumonia, Viral drug therapy
- Abstract
Background: SARS-CoV-2 enters cells by binding of its spike protein to angiotensin-converting enzyme 2 (ACE2). Angiotensin-converting enzyme inhibitors (ACEIs) or angiotensin II receptor blockers (ARBs) have been reported to increase ACE2 expression in animal models, and worse outcomes are reported in patients with co-morbidities commonly treated with these agents, leading to controversy during the COVID-19 pandemic over whether these drugs might be helpful or harmful., Methods: Animal, in vitro and clinical data relevant to the biology of the renin-angiotensin system (RAS), its interaction with the kallikrein-kinin system (KKS) and SARS-CoV-2, and clinical studies were reviewed., Findings and Interpretation: SARS-CoV-2 hijacks ACE2to invade and damage cells, downregulating ACE2, reducing its protective effects and exacerbating injurious Ang II effects. However, retrospective observational studies do not show higher risk of infection with ACEI or ARB use. Nevertheless, study of the RAS and KKS in the setting of coronaviral infection may yield therapeutic targets., (Copyright © 2020 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.