Seminars in Thrombosis and HemostasisThieme eJournals - The online journal service of the Thieme Publishing Group giving you access to some 130 medical and scientific journals.https://www.thieme-connect.com/products/ejournals/journal/10.1055/s-00000077Georg Thieme Verlag KGen© Georg Thieme Verlag KG Stuttgart · New YorkSeminars in Thrombosis and Hemostasis0094-61761098-9064© Georg Thieme Verlag KG Stuttgart · New YorkeJournals@thieme.de
  • Stability of Hemostasis Parameters in Whole Blood, Plasma, and Frozen Plasma: Literature Review and Recommendations of the SFTH (French Society of Thrombosis and Haemostasis)http://dx.doi.org/10.1055/s-0044-1788901Preanalytical sample management is critical for a proper assessment of hemostasis parameters, and may differ depending on prescribed tests or additional tests considered to be necessary after initial results. Although there is quite vast literature on this issue, the Working Group of the French Society of Thrombosis and Haemostasis (SFTH) deemed it necessary to make an in-depth literature review and propose recommendations for the proper handling of samples prior to hemostasis assays. This extensive assessment is accessible on-line in French at the SFTH website. Here, a more synthetic view of these recommendations is proposed, supported by easy-to-use tables. The latter respectively deal with the stability of whole blood or fresh plasma, frozen samples, and proper handling of samples forwarded on dry ice. Procedures are classified as recommended, acceptable, not conformed and lacking data. This work involved the retrieval of 125 references, first screened by a working group of 6 experts, then reviewed by 20 other experts in the field. The highly detailed conditions summarized in these tables will hopefully help hemostasis laboratories to secure the conditions recommended for sample collection and transportation. Moreover, as some conditions clearly lacked recommendations, this review can open new fields of investigation for hemostasis preanalytics.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1788901

    Preanalytical sample management is critical for a proper assessment of hemostasis parameters, and may differ depending on prescribed tests or additional tests considered to be necessary after initial results. Although there is quite vast literature on this issue, the Working Group of the French Society of Thrombosis and Haemostasis (SFTH) deemed it necessary to make an in-depth literature review and propose recommendations for the proper handling of samples prior to hemostasis assays. This extensive assessment is accessible on-line in French at the SFTH website. Here, a more synthetic view of these recommendations is proposed, supported by easy-to-use tables. The latter respectively deal with the stability of whole blood or fresh plasma, frozen samples, and proper handling of samples forwarded on dry ice. Procedures are classified as recommended, acceptable, not conformed and lacking data. This work involved the retrieval of 125 references, first screened by a working group of 6 experts, then reviewed by 20 other experts in the field. The highly detailed conditions summarized in these tables will hopefully help hemostasis laboratories to secure the conditions recommended for sample collection and transportation. Moreover, as some conditions clearly lacked recommendations, this review can open new fields of investigation for hemostasis preanalytics.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Stability of Hemostasis Parameters in Whole Blood, Plasma, and Frozen Plasma: Literature Review and Recommendations of the SFTH (French Society of Thrombosis and Haemostasis)Flaujac, ClaireDelassasseigne, CélineHurtaud-Roux, Marie-FrançoiseDelahousse, BenedicteBoissier, ElodieDesconclois, CélineDOI:10.1055/s-0044-1788901Semin Thromb Hemost ; : -2024-08-30T19:48:51+01:00Seminars in Thrombosis and Hemostasis2024-08-30T19:48:51+01:00eFirst
    Review Article
    10.1055/s-0044-1788901http://dx.doi.org/10.1055/s-0044-1788901
    Role of Platelets and Their Interaction with Immune Cells in Venous Thromboembolismhttp://dx.doi.org/10.1055/s-0044-1789022Venous thromboembolism (VTE) represents a significant global health challenge, ranking as the third leading cause of cardiovascular-related mortality. VTE pervades diverse clinical specialties, posing substantial risks to patient well-being and imposing considerable economic strains on health care systems. While platelets have long been recognized as pivotal players in hemostasis, emerging evidence underscores their multifaceted immune functions and their capacity to engage in crosstalk with other immune cells, such as neutrophils, thereby fostering immune-related thrombosis. Notably, investigations have elucidated the pivotal role of platelets in the pathogenesis of VTE. This review provides a comprehensive overview of platelet physiology, encompassing their activation, secretion dynamics, and implications in VTE. Moreover, it delineates the impact of platelet interactions with various immune cells on the initiation and progression of VTE, explores the correlation between platelet-related laboratory markers and VTE, and elucidates the role of platelets in thrombosis regression.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1789022

    Venous thromboembolism (VTE) represents a significant global health challenge, ranking as the third leading cause of cardiovascular-related mortality. VTE pervades diverse clinical specialties, posing substantial risks to patient well-being and imposing considerable economic strains on health care systems. While platelets have long been recognized as pivotal players in hemostasis, emerging evidence underscores their multifaceted immune functions and their capacity to engage in crosstalk with other immune cells, such as neutrophils, thereby fostering immune-related thrombosis. Notably, investigations have elucidated the pivotal role of platelets in the pathogenesis of VTE. This review provides a comprehensive overview of platelet physiology, encompassing their activation, secretion dynamics, and implications in VTE. Moreover, it delineates the impact of platelet interactions with various immune cells on the initiation and progression of VTE, explores the correlation between platelet-related laboratory markers and VTE, and elucidates the role of platelets in thrombosis regression.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Role of Platelets and Their Interaction with Immune Cells in Venous ThromboembolismZhang, ZhaoZhou, XianghuiZhou, XinCheng, ZhipengHu, YuDOI:10.1055/s-0044-1789022Semin Thromb Hemost ; : -2024-08-30T19:48:49+01:00Seminars in Thrombosis and Hemostasis2024-08-30T19:48:49+01:00eFirst
    Review Article
    10.1055/s-0044-1789022http://dx.doi.org/10.1055/s-0044-1789022
    Clinical, Laboratory, and Molecular Aspects of Factor VII Deficiencyhttp://dx.doi.org/10.1055/s-0044-1788792Congenital factor VII (FVII) deficiency, the most frequent among the recessively inherited disorders of blood coagulation, is characterized by a wide range of symptoms, from mild mucosal bleeds to life-threatening intracranial hemorrhage. Complete FVII deficiency may cause perinatal lethality. Clinically relevant thresholds of plasma levels are still uncertain, and modest differences in low FVII levels are associated with large differences in clinical phenotypes. Activated FVII (FVIIa) expresses its physiological protease activity only in a complex with tissue factor (TF), which triggers clotting at a very low concentration. Knowledge of the FVIIa–TF complex helps to interpret the clinical findings associated with low FVII activity as compared with other rare bleeding disorders and permits effective management, including prophylaxis, with recombinant FVIIa, which, however, displays a short half-life. Newly devised substitutive and nonsubstitutive treatments, characterized by extended half-life properties, may further improve the quality of life of patients. Genetic diagnosis has been performed in thousands of patients with FVII deficiency, and among the heterogeneous F7 mutations, mostly missense changes, several recurrent variants show geographical distribution and identity by descent. In the general population, common F7 polymorphisms explain a large proportion of FVII level variance in plasma through FVII-lowering effects. Their combination with pathogenic variants may impact on the frequent detection of FVII coagulant levels lower than normal, as well as on mild bleeding conditions. In the twenties of this century, 70 years after the first report of FVII deficiency, more than 200 studies/reports about FVII/FVII deficiency have been published, with thousands of FVII-deficient patients characterized all over the world.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1788792

    Congenital factor VII (FVII) deficiency, the most frequent among the recessively inherited disorders of blood coagulation, is characterized by a wide range of symptoms, from mild mucosal bleeds to life-threatening intracranial hemorrhage. Complete FVII deficiency may cause perinatal lethality. Clinically relevant thresholds of plasma levels are still uncertain, and modest differences in low FVII levels are associated with large differences in clinical phenotypes. Activated FVII (FVIIa) expresses its physiological protease activity only in a complex with tissue factor (TF), which triggers clotting at a very low concentration. Knowledge of the FVIIa–TF complex helps to interpret the clinical findings associated with low FVII activity as compared with other rare bleeding disorders and permits effective management, including prophylaxis, with recombinant FVIIa, which, however, displays a short half-life. Newly devised substitutive and nonsubstitutive treatments, characterized by extended half-life properties, may further improve the quality of life of patients. Genetic diagnosis has been performed in thousands of patients with FVII deficiency, and among the heterogeneous F7 mutations, mostly missense changes, several recurrent variants show geographical distribution and identity by descent. In the general population, common F7 polymorphisms explain a large proportion of FVII level variance in plasma through FVII-lowering effects. Their combination with pathogenic variants may impact on the frequent detection of FVII coagulant levels lower than normal, as well as on mild bleeding conditions. In the twenties of this century, 70 years after the first report of FVII deficiency, more than 200 studies/reports about FVII/FVII deficiency have been published, with thousands of FVII-deficient patients characterized all over the world.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Clinical, Laboratory, and Molecular Aspects of Factor VII DeficiencyBernardi, FrancescoMariani, GuglielmoDOI:10.1055/s-0044-1788792Semin Thromb Hemost ; : -2024-08-29T09:45:43+01:00Seminars in Thrombosis and Hemostasis2024-08-29T09:45:43+01:00eFirst
    Review Article
    10.1055/s-0044-1788792http://dx.doi.org/10.1055/s-0044-1788792
    Clinical, Laboratory Aspects and Management of Factor X Deficiencyhttp://dx.doi.org/10.1055/s-0044-1789595Coagulation factor X (FX), originally named Stuart–Prower factor, plays a pivotal role in the coagulation cascade, activating thrombin to promote platelet plug formation and prevent excess blood loss. Genetic variants in F10 may lead to FX deficiency and to impaired coagulation. FX variants are phenotypically classified as being type I, with the concomitant reduction of FX coagulant activity and FX antigen levels or type II, corresponding to a reduction in activity with normal antigen plasma levels. Patients affected with FX deficiency tend to be one of the most seriously affected among those with rare bleeding disorders. They show a variable bleeding tendency strongly associated with FX coagulant activity levels in plasma and may present, in the severe form of the deficiency, life-threatening symptoms such as gastrointestinal and umbilical stump bleeding and intracranial hemorrhages or central nervous system bleeding. Treatment of FX deficiency was originally based on the replacement of the missing factor using fresh frozen plasma, cryoprecipitate and prothrombin complex concentrates; however, a plasma-derived concentrate, shown to be safe and effective in clinical trials, is now available. In addition, novel nonreplacement therapy such as small interference RNA, gene therapy, drug repurposing, and gene editing may also represent novel therapeutic approaches for FX deficiency, but further, much focused studies are needed before considering this emerging therapy in such patients.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1789595

    Coagulation factor X (FX), originally named Stuart–Prower factor, plays a pivotal role in the coagulation cascade, activating thrombin to promote platelet plug formation and prevent excess blood loss. Genetic variants in F10 may lead to FX deficiency and to impaired coagulation. FX variants are phenotypically classified as being type I, with the concomitant reduction of FX coagulant activity and FX antigen levels or type II, corresponding to a reduction in activity with normal antigen plasma levels. Patients affected with FX deficiency tend to be one of the most seriously affected among those with rare bleeding disorders. They show a variable bleeding tendency strongly associated with FX coagulant activity levels in plasma and may present, in the severe form of the deficiency, life-threatening symptoms such as gastrointestinal and umbilical stump bleeding and intracranial hemorrhages or central nervous system bleeding. Treatment of FX deficiency was originally based on the replacement of the missing factor using fresh frozen plasma, cryoprecipitate and prothrombin complex concentrates; however, a plasma-derived concentrate, shown to be safe and effective in clinical trials, is now available. In addition, novel nonreplacement therapy such as small interference RNA, gene therapy, drug repurposing, and gene editing may also represent novel therapeutic approaches for FX deficiency, but further, much focused studies are needed before considering this emerging therapy in such patients.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Clinical, Laboratory Aspects and Management of Factor X DeficiencyMenegatti, MarziaPeyvandi, FloraDOI:10.1055/s-0044-1789595Semin Thromb Hemost ; : -2024-08-29T09:45:41+01:00Seminars in Thrombosis and Hemostasis2024-08-29T09:45:41+01:00eFirst
    Review Article
    10.1055/s-0044-1789595http://dx.doi.org/10.1055/s-0044-1789595
    Clinical, Laboratory, Molecular, and Reproductive Aspects of Combined Deficiency of Factors V and VIIIhttp://dx.doi.org/10.1055/s-0044-1789019Congenital combined deficiency of factor V (FV) and factor VIII (FVIII; F5F8D, OMIM 227300) is a rare hereditary coagulopathy and accounts for approximately 3% of cases of rare coagulation disorders. The prevalence of this disease in the general population is estimated to be 1:1,000,000 and is significantly higher in regions where consanguineous marriages are permitted, such as the Mideast and South Asia. The disease has an autosomal recessive mode of inheritance and therefore occurs with an equal incidence among males and females. Heterozygous mutation carriers usually do not have clinical manifestations. The molecular basis of this disease differs from that of stand-alone congenital deficiencies of FVIII and FV. F5F8D is caused by mutations in either LMAN1 or MCFD2, which encode components of a cargo receptor complex for endoplasmic reticulum to Golgi transport of FV and FVIII, leading to defects in an intracellular transport pathway shared by these two coagulation factors. Congenital combined deficiency of FV and FVIII is characterized by decreased activities of both FV and FVIII in plasma, usually to 5 to 30% of normal. Clinical manifestations in most cases are represented by mild or moderate hemorrhagic syndrome. The simultaneous decreases of two coagulation factors present complications in the diagnosis and management of the disease. In female patients, the disease requires a special approach for family planning, pregnancy management, and parturition. This review summarizes recent progress in clinical, laboratory, and molecular understanding of this disorder.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1789019

    Congenital combined deficiency of factor V (FV) and factor VIII (FVIII; F5F8D, OMIM 227300) is a rare hereditary coagulopathy and accounts for approximately 3% of cases of rare coagulation disorders. The prevalence of this disease in the general population is estimated to be 1:1,000,000 and is significantly higher in regions where consanguineous marriages are permitted, such as the Mideast and South Asia. The disease has an autosomal recessive mode of inheritance and therefore occurs with an equal incidence among males and females. Heterozygous mutation carriers usually do not have clinical manifestations. The molecular basis of this disease differs from that of stand-alone congenital deficiencies of FVIII and FV. F5F8D is caused by mutations in either LMAN1 or MCFD2, which encode components of a cargo receptor complex for endoplasmic reticulum to Golgi transport of FV and FVIII, leading to defects in an intracellular transport pathway shared by these two coagulation factors. Congenital combined deficiency of FV and FVIII is characterized by decreased activities of both FV and FVIII in plasma, usually to 5 to 30% of normal. Clinical manifestations in most cases are represented by mild or moderate hemorrhagic syndrome. The simultaneous decreases of two coagulation factors present complications in the diagnosis and management of the disease. In female patients, the disease requires a special approach for family planning, pregnancy management, and parturition. This review summarizes recent progress in clinical, laboratory, and molecular understanding of this disorder.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Clinical, Laboratory, Molecular, and Reproductive Aspects of Combined Deficiency of Factors V and VIIIYakovleva, ElenaZhang, BinDOI:10.1055/s-0044-1789019Semin Thromb Hemost ; : -2024-08-29T09:45:42+01:00Seminars in Thrombosis and Hemostasis2024-08-29T09:45:42+01:00eFirst
    Review Article
    10.1055/s-0044-1789019http://dx.doi.org/10.1055/s-0044-1789019
    Clinical, Laboratory, and Molecular Aspects of Factor V Deficiencyhttp://dx.doi.org/10.1055/s-0044-1789021Factor V (FV) is a glycoprotein that plays a pivotal role in hemostasis, being involved in coagulant and anticoagulant pathways. Congenital FV deficiency is a rare bleeding disorder with an incidence of 1 per million live births, considering the most severe homozygous form. FV deficiency is diagnosed using routine coagulation tests and FV activity assays. Several mutations, including missense, nonsense, and frameshift, have been detected in the F5 gene. Clinical symptoms are variable, ranging from mild ecchymoses and mucosal bleeding to life-threatening intracranial hemorrhage. The mainstay of treatment includes fresh-frozen plasma, preferentially virus-inactivated. In this narrative review, we provide an update of the main laboratory, molecular, clinical, and therapeutic features of inherited FV deficiency.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1789021

    Factor V (FV) is a glycoprotein that plays a pivotal role in hemostasis, being involved in coagulant and anticoagulant pathways. Congenital FV deficiency is a rare bleeding disorder with an incidence of 1 per million live births, considering the most severe homozygous form. FV deficiency is diagnosed using routine coagulation tests and FV activity assays. Several mutations, including missense, nonsense, and frameshift, have been detected in the F5 gene. Clinical symptoms are variable, ranging from mild ecchymoses and mucosal bleeding to life-threatening intracranial hemorrhage. The mainstay of treatment includes fresh-frozen plasma, preferentially virus-inactivated. In this narrative review, we provide an update of the main laboratory, molecular, clinical, and therapeutic features of inherited FV deficiency.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Clinical, Laboratory, and Molecular Aspects of Factor V DeficiencyFranchini, MassimoFocosi, DanieleDOI:10.1055/s-0044-1789021Semin Thromb Hemost ; : -2024-08-27T13:09:55+01:00Seminars in Thrombosis and Hemostasis2024-08-27T13:09:55+01:00eFirst
    Review Article
    10.1055/s-0044-1789021http://dx.doi.org/10.1055/s-0044-1789021
    Platelet-Type von Willebrand Disease: Complex Pathophysiology and Insights on Novel Therapeutic and Diagnostic Strategieshttp://dx.doi.org/10.1055/s-0044-1789183von Willebrand disease (VWD) is the most common well-studied genetic bleeding disorder worldwide. Much less is known about platelet-type VWD (PT-VWD), a rare platelet function defect, and a “nonidentical” twin bleeding phenotype to type 2B VWD (2B-VWD). Rather than a defect in the von Willebrand factor (VWF) gene, PT-VWD is caused by a platelet GP1BA mutation leading to a hyperaffinity of the glycoprotein Ibα (GPIbα) platelet surface receptor for VWF, and thus increased platelet clearing and high-molecular-weight VWF multimer elimination. Nine GP1BA gene mutations are known. It is historically believed that this enhanced binding was enabled by the β-switch region of GPIbα adopting an extended β-hairpin form. Recent evidence suggests the pathological conformation that destabilizes the compact triangular form of the R-loop—the GPIbα protein's region for VWF binding. PT-VWD is often misdiagnosed as 2B-VWD, even the though distinction between the two is crucial for proper treatment, as the former requires platelet transfusions, while the latter requires VWF/FVIII concentrate administration. Nevertheless, these PT-VWD treatments remain unsatisfactory, owing to their high cost, low availability, risk of alloimmunity, and the need to carefully balance platelet administration. Antibodies such as 6B4 remain undependable as an alternative therapy due to their questionable efficacy and high costs for this purpose. On the other hand, synthetic peptide therapeutics developed with In-Silico Protein Synthesizer to disrupt the association between GPIbα and VWF show preliminary promise as a therapy based on in vitro experiments. Such peptides could serve as an effective diagnostic technology for discriminating between 2B-VWD and PT-VWD, or potentially all forms of VWD, based on their high specificity. This field is rapidly growing and the current review sheds light on the complex pathology and some novel potential therapeutic and diagnostic strategies.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1789183

    von Willebrand disease (VWD) is the most common well-studied genetic bleeding disorder worldwide. Much less is known about platelet-type VWD (PT-VWD), a rare platelet function defect, and a “nonidentical” twin bleeding phenotype to type 2B VWD (2B-VWD). Rather than a defect in the von Willebrand factor (VWF) gene, PT-VWD is caused by a platelet GP1BA mutation leading to a hyperaffinity of the glycoprotein Ibα (GPIbα) platelet surface receptor for VWF, and thus increased platelet clearing and high-molecular-weight VWF multimer elimination. Nine GP1BA gene mutations are known. It is historically believed that this enhanced binding was enabled by the β-switch region of GPIbα adopting an extended β-hairpin form. Recent evidence suggests the pathological conformation that destabilizes the compact triangular form of the R-loop—the GPIbα protein's region for VWF binding. PT-VWD is often misdiagnosed as 2B-VWD, even the though distinction between the two is crucial for proper treatment, as the former requires platelet transfusions, while the latter requires VWF/FVIII concentrate administration. Nevertheless, these PT-VWD treatments remain unsatisfactory, owing to their high cost, low availability, risk of alloimmunity, and the need to carefully balance platelet administration. Antibodies such as 6B4 remain undependable as an alternative therapy due to their questionable efficacy and high costs for this purpose. On the other hand, synthetic peptide therapeutics developed with In-Silico Protein Synthesizer to disrupt the association between GPIbα and VWF show preliminary promise as a therapy based on in vitro experiments. Such peptides could serve as an effective diagnostic technology for discriminating between 2B-VWD and PT-VWD, or potentially all forms of VWD, based on their high specificity. This field is rapidly growing and the current review sheds light on the complex pathology and some novel potential therapeutic and diagnostic strategies.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Platelet-Type von Willebrand Disease: Complex Pathophysiology and Insights on Novel Therapeutic and Diagnostic StrategiesFu, AnneKazmirchuk, Thomas D.D.Bradbury-Jost, CalvinGolshani, AshkanOthman, MahaDOI:10.1055/s-0044-1789183Semin Thromb Hemost ; : -2024-08-27T13:09:52+01:00Seminars in Thrombosis and Hemostasis2024-08-27T13:09:52+01:00eFirst
    Review Article
    10.1055/s-0044-1789183http://dx.doi.org/10.1055/s-0044-1789183
    Platelets in Vascular Calcification: A Comprehensive Review of Platelet-Derived Extracellular Vesicles, Protein Interactions, Platelet Function Indices, and their Impact on Cellular Crosstalkhttp://dx.doi.org/10.1055/s-0044-1789023Vascular calcification (VC) commonly accompanies the development of atherosclerosis, defined by the accumulation of calcium in the arterial wall, potentially leading to stroke and myocardial infarction. Severe and unevenly distributed calcification poses challenges for interventional procedures, elevating the risks of vascular dissection, acute vascular occlusion, restenosis, and other major adverse cardiovascular events. Platelets promote the development of atherosclerosis by secreting various inflammatory mediators, regulating cell migration, aggregation, adhesion, and initiating and expanding inflammatory responses. There is emerging evidence that platelets play a direct role in VC; however, this novel concept has not yet been critically assessed. This review describes the intricate mechanisms by which platelets promote VC, focusing on three key aspects and the potential opportunities for their therapeutic targeting: extracellular vesicles, platelet-regulatory proteins, and indices related to platelet function.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1789023

    Vascular calcification (VC) commonly accompanies the development of atherosclerosis, defined by the accumulation of calcium in the arterial wall, potentially leading to stroke and myocardial infarction. Severe and unevenly distributed calcification poses challenges for interventional procedures, elevating the risks of vascular dissection, acute vascular occlusion, restenosis, and other major adverse cardiovascular events. Platelets promote the development of atherosclerosis by secreting various inflammatory mediators, regulating cell migration, aggregation, adhesion, and initiating and expanding inflammatory responses. There is emerging evidence that platelets play a direct role in VC; however, this novel concept has not yet been critically assessed. This review describes the intricate mechanisms by which platelets promote VC, focusing on three key aspects and the potential opportunities for their therapeutic targeting: extracellular vesicles, platelet-regulatory proteins, and indices related to platelet function.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Platelets in Vascular Calcification: A Comprehensive Review of Platelet-Derived Extracellular Vesicles, Protein Interactions, Platelet Function Indices, and their Impact on Cellular CrosstalkHe, YiZhang, QiongyuePan, LinaYang, HaoLiu, TaoBei, JunjiePeter, KarlheinzHu, HouyuanDOI:10.1055/s-0044-1789023Semin Thromb Hemost ; : -2024-08-27T13:09:53+01:00Seminars in Thrombosis and Hemostasis2024-08-27T13:09:53+01:00eFirst
    Review Article
    10.1055/s-0044-1789023http://dx.doi.org/10.1055/s-0044-1789023
    Discordant High Activated Partial Thromboplastin Time Relative to Anti-Xa Values in Hospitalized Patients is an Independent Risk Factor for Increased 30-day Mortalityhttp://dx.doi.org/10.1055/s-0044-1789020 Background The activated partial thromboplastin time (aPTT) and anti-factor-Xa levels (anti-Xa) are both used to monitor patients on unfractionated heparin. Our previous study demonstrated that patients with discordant high aPTT relative to anti-Xa had higher rates of mortality and bleeding events. Objective To determine if underlying patient characteristics drive both discordance and adverse outcomes or if discordance is an independent risk factor to adverse outcomes. Methods We analyzed all patients hospitalized at the Stanford Hospital between January 2011 and December 2019 who had simultaneous aPTT and anti-Xa levels performed. From the electronic medical record, we extracted and analyzed 51 patient features including baseline coagulation laboratory results, demographics, values of other common laboratories (basic metabolic panel, complete blood count, etc.), diagnostic procedures, medications, and death. Results A total of 17,728 patients had 78,701 paired aPTT and anti-Xa levels. Patients with discordant aPTT and anti-Xa where aPTT (seconds) was elevated beyond the expected therapeutic range had a higher 30-day mortality (odds ratio [OR]: 2.16, 95% confidence interval [CI]: 1.78–2.63, p < 0.001). Sectioning the patients based on the degree of discordance and whether aPTT or anti-Xa were signaling excess anticoagulation, we found those with an elevated aPTT discordant to their anti-Xa level had the highest odds of death (OR: 2.46, 95% CI: 1.99–3.10) compared with the concordant group. This finding was still present after controlling for patient comorbidity and other laboratory results at hospital admission. Conclusion After controlling for patient features strongly associated with increased mortality in heparinized patients, we identified that the discordant pattern of high aPTT to anti-Xa served as an independent predictor of 30-day all-cause mortality, with a higher degree of discordance associated with increased odds of 30-day mortality.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1789020

    Background The activated partial thromboplastin time (aPTT) and anti-factor-Xa levels (anti-Xa) are both used to monitor patients on unfractionated heparin. Our previous study demonstrated that patients with discordant high aPTT relative to anti-Xa had higher rates of mortality and bleeding events. Objective To determine if underlying patient characteristics drive both discordance and adverse outcomes or if discordance is an independent risk factor to adverse outcomes. Methods We analyzed all patients hospitalized at the Stanford Hospital between January 2011 and December 2019 who had simultaneous aPTT and anti-Xa levels performed. From the electronic medical record, we extracted and analyzed 51 patient features including baseline coagulation laboratory results, demographics, values of other common laboratories (basic metabolic panel, complete blood count, etc.), diagnostic procedures, medications, and death. Results A total of 17,728 patients had 78,701 paired aPTT and anti-Xa levels. Patients with discordant aPTT and anti-Xa where aPTT (seconds) was elevated beyond the expected therapeutic range had a higher 30-day mortality (odds ratio [OR]: 2.16, 95% confidence interval [CI]: 1.78–2.63, p < 0.001). Sectioning the patients based on the degree of discordance and whether aPTT or anti-Xa were signaling excess anticoagulation, we found those with an elevated aPTT discordant to their anti-Xa level had the highest odds of death (OR: 2.46, 95% CI: 1.99–3.10) compared with the concordant group. This finding was still present after controlling for patient comorbidity and other laboratory results at hospital admission. Conclusion After controlling for patient features strongly associated with increased mortality in heparinized patients, we identified that the discordant pattern of high aPTT to anti-Xa served as an independent predictor of 30-day all-cause mortality, with a higher degree of discordance associated with increased odds of 30-day mortality.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Discordant High Activated Partial Thromboplastin Time Relative to Anti-Xa Values in Hospitalized Patients is an Independent Risk Factor for Increased 30-day MortalityJin, JingGummidipundi, SantoshHsu, JoeSharifi, HushamBoothroyd, DerekKrishnan, AnandiZehnder, James L.DOI:10.1055/s-0044-1789020Semin Thromb Hemost ; : -2024-08-27T13:09:57+01:00Seminars in Thrombosis and Hemostasis2024-08-27T13:09:57+01:00eFirst
    Review Article
    10.1055/s-0044-1789020http://dx.doi.org/10.1055/s-0044-1789020
    Bernard–Soulier Syndrome: A Review of Epidemiology, Molecular Pathology, Clinical Features, Laboratory Diagnosis, and Therapeutic Managementhttp://dx.doi.org/10.1055/s-0044-1789184Bernard–Soulier syndrome (BSS) is an inherited platelet function disorder caused by mutations in the genes that encode the glycoprotein (GP) Ibα and GPIbβ subunits, as well as the GPIX subunit in the GPIbIX complex, which is located on the platelet surface and has roles in platelet adhesion and activation. Patients with autosomal recessively inherited biallelic BSS have a homozygous or compound heterozygous expression in the GPIbα, GPIbβ, and GPIX subunits of the GPIbIX complex. Patients with autosomal dominantly inherited monoallelic BSS have a heterozygous expression in only the GPIbα and GPIbβ subunits of the GPIbIX complex. To date, no BSS mutations in the GP5 gene have been reported. Patients with biallelic form are usually diagnosed at a young age, typically with mucocutaneous bleeding, whereas monoallelic forms are generally identified later in life and are frequently misdiagnosed with immune thrombocytopenic purpura (ITP). In biallelic BSS, giant platelets in the peripheral blood smear, absence of ristocetin-induced platelet aggregation (RIPA) using light transmission aggregometry (LTA), and complete loss of GPIbIX complex in flow cytometry are observed, whereas in monoallelic forms, genetic diagnosis is recommended due to the presence of large platelets in the peripheral blood smear, decreased or normal RIPA response in LTA, and partial loss or normal GPIbIX complex in flow cytometry. Platelet transfusion is the main therapy but recombinant factor VIIa is advised in alloimmunized patients, and allogeneic stem cell transplantation is suggested in refractory cases. Antifibrinolytics and oral contraceptives are utilized as supplementary treatments. Finally, differentiation from ITP is critical due to differences in management. Thus, BSS should be kept in mind in the presence of individuals with chronic persistent thrombocytopenia, positive family history, unresponsive ITP treatment, macrothrombocytopenia, and absence of RIPA response.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1789184

    Bernard–Soulier syndrome (BSS) is an inherited platelet function disorder caused by mutations in the genes that encode the glycoprotein (GP) Ibα and GPIbβ subunits, as well as the GPIX subunit in the GPIbIX complex, which is located on the platelet surface and has roles in platelet adhesion and activation. Patients with autosomal recessively inherited biallelic BSS have a homozygous or compound heterozygous expression in the GPIbα, GPIbβ, and GPIX subunits of the GPIbIX complex. Patients with autosomal dominantly inherited monoallelic BSS have a heterozygous expression in only the GPIbα and GPIbβ subunits of the GPIbIX complex. To date, no BSS mutations in the GP5 gene have been reported. Patients with biallelic form are usually diagnosed at a young age, typically with mucocutaneous bleeding, whereas monoallelic forms are generally identified later in life and are frequently misdiagnosed with immune thrombocytopenic purpura (ITP). In biallelic BSS, giant platelets in the peripheral blood smear, absence of ristocetin-induced platelet aggregation (RIPA) using light transmission aggregometry (LTA), and complete loss of GPIbIX complex in flow cytometry are observed, whereas in monoallelic forms, genetic diagnosis is recommended due to the presence of large platelets in the peripheral blood smear, decreased or normal RIPA response in LTA, and partial loss or normal GPIbIX complex in flow cytometry. Platelet transfusion is the main therapy but recombinant factor VIIa is advised in alloimmunized patients, and allogeneic stem cell transplantation is suggested in refractory cases. Antifibrinolytics and oral contraceptives are utilized as supplementary treatments. Finally, differentiation from ITP is critical due to differences in management. Thus, BSS should be kept in mind in the presence of individuals with chronic persistent thrombocytopenia, positive family history, unresponsive ITP treatment, macrothrombocytopenia, and absence of RIPA response.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Bernard–Soulier Syndrome: A Review of Epidemiology, Molecular Pathology, Clinical Features, Laboratory Diagnosis, and Therapeutic ManagementKaya, ZühreDOI:10.1055/s-0044-1789184Semin Thromb Hemost ; : -2024-08-27T13:09:56+01:00Seminars in Thrombosis and Hemostasis2024-08-27T13:09:56+01:00eFirst
    Review Article
    10.1055/s-0044-1789184http://dx.doi.org/10.1055/s-0044-1789184
    Hereditary Thrombophilia: A Case of Subacute Pulmonary Embolism in a 68-Year-Old Female with a Mutation in the PROC Genehttp://dx.doi.org/10.1055/s-0044-1788304Semin Thromb Hemost
    DOI: 10.1055/s-0044-1788304



    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Full text

    ]]>
    Hereditary Thrombophilia: A Case of Subacute Pulmonary Embolism in a 68-Year-Old Female with a Mutation in the PROC GeneChen, ZiyangWu, HualanXu, YuanGu, TaoLiu, DepingLan, MingDOI:10.1055/s-0044-1788304Semin Thromb Hemost ; : -2024-08-16T09:14:33+01:00Seminars in Thrombosis and Hemostasis2024-08-16T09:14:33+01:00eFirst
    Letter to the Editor
    10.1055/s-0044-1788304http://dx.doi.org/10.1055/s-0044-1788304
    Clinical, Laboratory, and Molecular Aspects of Congenital Fibrinogen Disordershttp://dx.doi.org/10.1055/s-0044-1788898Congenital fibrinogen disorders (CFDs) include afibrinogenemia, hypofibrinogenemia, dysfibrinogenemia, and hypodysfibrinogenemia. The fibrinogen levels, the clinical features, and the genotype define several sub-types, each with specific biological and clinical issues. The diagnosis of CFDs is based on the measurement of activity and antigen fibrinogen levels as well as on the genotype. While relatively easy in quantitative fibrinogen disorders, the diagnosis can be more challenging in qualitative fibrinogen disorders depending on the reagents and methods used, and the underlying fibrinogen variants. Overall, quantitative and qualitative fibrinogen defects lead to a decrease in clottability, and usually in a bleeding tendency. The severity of the bleeding phenotype is moreover related to the concentration of fibrinogen. Paradoxically, patients with CFDs are also at risk of thrombotic events. The impact of the causative mutation on the structure and the fibrinogen level is one of the determinants of the thrombotic profile. Given the major role of fibrinogen in pregnancy, women with CFDs are particularly at risk of obstetrical adverse outcomes. The study of the fibrin clot properties can help to define the impact of fibrinogen disorders on the fibrin network. The development of next generation sequencing now allows the identification of genetic modifiers able to influence the global hemostasis balance in CFDs. Their integration in the assessment of the patient risk on an individual scale is an important step toward precision medicine in patients with such a heterogeneous clinical course.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1788898

    Congenital fibrinogen disorders (CFDs) include afibrinogenemia, hypofibrinogenemia, dysfibrinogenemia, and hypodysfibrinogenemia. The fibrinogen levels, the clinical features, and the genotype define several sub-types, each with specific biological and clinical issues. The diagnosis of CFDs is based on the measurement of activity and antigen fibrinogen levels as well as on the genotype. While relatively easy in quantitative fibrinogen disorders, the diagnosis can be more challenging in qualitative fibrinogen disorders depending on the reagents and methods used, and the underlying fibrinogen variants. Overall, quantitative and qualitative fibrinogen defects lead to a decrease in clottability, and usually in a bleeding tendency. The severity of the bleeding phenotype is moreover related to the concentration of fibrinogen. Paradoxically, patients with CFDs are also at risk of thrombotic events. The impact of the causative mutation on the structure and the fibrinogen level is one of the determinants of the thrombotic profile. Given the major role of fibrinogen in pregnancy, women with CFDs are particularly at risk of obstetrical adverse outcomes. The study of the fibrin clot properties can help to define the impact of fibrinogen disorders on the fibrin network. The development of next generation sequencing now allows the identification of genetic modifiers able to influence the global hemostasis balance in CFDs. Their integration in the assessment of the patient risk on an individual scale is an important step toward precision medicine in patients with such a heterogeneous clinical course.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Clinical, Laboratory, and Molecular Aspects of Congenital Fibrinogen DisordersCasini, AlessandroMoerloose, Philippe deNeerman-Arbez, MargueriteDOI:10.1055/s-0044-1788898Semin Thromb Hemost ; : -2024-08-16T09:14:37+01:00Seminars in Thrombosis and Hemostasis2024-08-16T09:14:37+01:00eFirst
    Review Article
    10.1055/s-0044-1788898http://dx.doi.org/10.1055/s-0044-1788898
    Catastrophic Thrombosis: A Narrative Reviewhttp://dx.doi.org/10.1055/s-0044-1788790Catastrophic thrombosis is a severe condition characterized by a hypercoagulable tendency, leading to multiple thromboembolic events in different blood vessels, usually within a short timeframe. Several conditions have been associated with the development of catastrophic thrombosis, including the catastrophic antiphospholipid syndrome, thrombotic anti-platelet factor 4 immune disorders, thrombotic microangiopathies, cancers, the hyper-eosinophilic syndrome, pregnancy, infections, trauma, and drugs. Thrombotic storm represents a medical emergency whose management represents a serious challenge for physicians. Besides the prompt start of anticoagulation, a patient's prognosis depends on early recognition and possible treatment of the underlying condition. In this narrative review, we summarize the main characteristics of catastrophic thrombosis, analyzing the various conditions triggering such life-threatening complication. Finally, an algorithm with the diagnostic workup and the initial management of patients with catastrophic thrombosis is presented.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1788790

    Catastrophic thrombosis is a severe condition characterized by a hypercoagulable tendency, leading to multiple thromboembolic events in different blood vessels, usually within a short timeframe. Several conditions have been associated with the development of catastrophic thrombosis, including the catastrophic antiphospholipid syndrome, thrombotic anti-platelet factor 4 immune disorders, thrombotic microangiopathies, cancers, the hyper-eosinophilic syndrome, pregnancy, infections, trauma, and drugs. Thrombotic storm represents a medical emergency whose management represents a serious challenge for physicians. Besides the prompt start of anticoagulation, a patient's prognosis depends on early recognition and possible treatment of the underlying condition. In this narrative review, we summarize the main characteristics of catastrophic thrombosis, analyzing the various conditions triggering such life-threatening complication. Finally, an algorithm with the diagnostic workup and the initial management of patients with catastrophic thrombosis is presented.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Catastrophic Thrombosis: A Narrative ReviewFranchini, MassimoFocosi, DanielePezzo, Mario PiergiulioMannucci, Pier MannuccioDOI:10.1055/s-0044-1788790Semin Thromb Hemost ; : -2024-08-16T09:14:38+01:00Seminars in Thrombosis and Hemostasis2024-08-16T09:14:38+01:00eFirst
    Review Article
    10.1055/s-0044-1788790http://dx.doi.org/10.1055/s-0044-1788790
    Comparison of Thrombophilia Assay Results for the International Society on Thrombosis and Haemostasis Scientific and Standardization Committee Plasma Standard from Different External Quality Assessment Providers—for the External Quality Assurance in Thrombosis and Haemostasis Grouphttp://dx.doi.org/10.1055/s-0044-1788791External quality assessment (EQA) is used to evaluate laboratory performance in tests of hemostasis; however, some esoteric tests are performed by too few centers in any one EQA program to allow valid statistical assessment. To explore the feasibility of pooling data from several EQA providers, an exercise was carried out by the External Quality Assurance in Thrombosis and Haemostasis group, using the International Society on Thrombosis and Haemostasis Scientific and Standardization Committee (SSC) plasma standard for thrombophilia screening assays. Six EQA providers took part in this exercise, distributing the SSC plasma standard as a “blinded” sample to participants for thrombophilia tests between November 2020 and December 2021. Data were collected by each provider, anonymized, and pooled for analysis. Results were analyzed as overall results from each EQA provider, and by kit/method-specific comparisons of data from all providers pooled together. For each parameter, median results and range were determined. Over 1,250 sets of data were returned in the six EQA programs. The overall medians (all data pooled) were <4% of the assigned values for each parameter with the exception of protein C activity by clot-based assay. Method-related differences in median results were observed for free protein S antigen and protein S activity—a pattern seen across data from the different EQA providers. Antithrombin antigen results reported in mg/dL provided an example where small numbers of results for a single EQA provider may be supplemented by pooling data from multiple providers with good agreement seen among results reported by the different EQA providers. This study demonstrated that a multicenter EQA provider collaboration can be carried out and demonstrated benefit for assays with smaller number of participants. In addition, results showed good agreement with the assigned values of the SSC plasma standard. Further exercises for tests performed by only small numbers of laboratories can be planned.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1788791

    External quality assessment (EQA) is used to evaluate laboratory performance in tests of hemostasis; however, some esoteric tests are performed by too few centers in any one EQA program to allow valid statistical assessment. To explore the feasibility of pooling data from several EQA providers, an exercise was carried out by the External Quality Assurance in Thrombosis and Haemostasis group, using the International Society on Thrombosis and Haemostasis Scientific and Standardization Committee (SSC) plasma standard for thrombophilia screening assays. Six EQA providers took part in this exercise, distributing the SSC plasma standard as a “blinded” sample to participants for thrombophilia tests between November 2020 and December 2021. Data were collected by each provider, anonymized, and pooled for analysis. Results were analyzed as overall results from each EQA provider, and by kit/method-specific comparisons of data from all providers pooled together. For each parameter, median results and range were determined. Over 1,250 sets of data were returned in the six EQA programs. The overall medians (all data pooled) were <4% of the assigned values for each parameter with the exception of protein C activity by clot-based assay. Method-related differences in median results were observed for free protein S antigen and protein S activity—a pattern seen across data from the different EQA providers. Antithrombin antigen results reported in mg/dL provided an example where small numbers of results for a single EQA provider may be supplemented by pooling data from multiple providers with good agreement seen among results reported by the different EQA providers. This study demonstrated that a multicenter EQA provider collaboration can be carried out and demonstrated benefit for assays with smaller number of participants. In addition, results showed good agreement with the assigned values of the SSC plasma standard. Further exercises for tests performed by only small numbers of laboratories can be planned.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Comparison of Thrombophilia Assay Results for the International Society on Thrombosis and Haemostasis Scientific and Standardization Committee Plasma Standard from Different External Quality Assessment Providers—for the External Quality Assurance in Thrombosis and Haemostasis GroupJennings, IanMeijer, PietArunachalam, SandyaMarlar, Richard A.Olson, John D.Zantek, Nicole D.Bon, ChantalDean, ElysseHollestelle, Martine J.Meley, RolandPlumhoff, Elizabeth A.Reilly-Stitt, ChristopherSalazar, EricSmock, Kristi J.Spannagl, MichaelWalker, Isobel D.DOI:10.1055/s-0044-1788791Semin Thromb Hemost ; : -2024-08-16T09:14:36+01:00Seminars in Thrombosis and Hemostasis2024-08-16T09:14:36+01:00eFirst
    Review Article
    10.1055/s-0044-1788791http://dx.doi.org/10.1055/s-0044-1788791
    Heparin and Posttraumatic Stress Disorderhttp://dx.doi.org/10.1055/s-0044-1788697Semin Thromb Hemost
    DOI: 10.1055/s-0044-1788697



    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Full text

    ]]>
    Heparin and Posttraumatic Stress DisorderLippi, GiuseppeFavaloro, Emmanuel J.DOI:10.1055/s-0044-1788697Semin Thromb Hemost ; : -2024-07-24T20:09:02+01:00Seminars in Thrombosis and Hemostasis2024-07-24T20:09:02+01:00eFirst
    Commentary
    10.1055/s-0044-1788697http://dx.doi.org/10.1055/s-0044-1788697
    Heparin-Like Anticoagulant: First Case Described in a Metastatic Biliopancreatic Cancerhttp://dx.doi.org/10.1055/s-0044-1788734Semin Thromb Hemost
    DOI: 10.1055/s-0044-1788734



    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Full text

    ]]>
    Heparin-Like Anticoagulant: First Case Described in a Metastatic Biliopancreatic CancerRaimondi, RosanaAguirre, SofiaMonserrat, ValeriaPons, SilvinaContardo, DamianScazziota, AlejandraDOI:10.1055/s-0044-1788734Semin Thromb Hemost ; : -2024-07-24T20:09:00+01:00Seminars in Thrombosis and Hemostasis2024-07-24T20:09:00+01:00eFirst
    Letter to the Editor
    10.1055/s-0044-1788734http://dx.doi.org/10.1055/s-0044-1788734
    Antiphospholipid Antibodies in Mental Disordershttp://dx.doi.org/10.1055/s-0044-1788696Thrombotic events striking the central nervous system are clinical criteria for the antiphospholipid syndrome (APS). Besides these, neuropsychiatric non-APS criteria manifestations are increasingly described in patients with persistently positive antiphospholipid antibodies (aPL). Among these are psychiatric manifestations. Animal models mainly describe hyperactive behavior and anxiety associated with hippocampal abnormalities. Cases of associations with psychosis, mood disorders, bipolarity, anxiety, obsessive–compulsive behavior, and depression have been reported but are still rare. Systematic human clinical association studies are concordant with a risk of psychosis, depression (simple to major), and anxiety disorders, but these are limited and of inconstant methodological quality. Brain imaging in patients, also insufficiently investigated, shows early signs of hypoperfusion and of subtle diffuse white matter changes compatible with an alteration of the axonal structure and changes in the myelin sheath. Direct interactions of aPL with the brain cells, both on cell lines and on animal and human brain biopsies, targeting both glial cells, astrocytes, and neurons, can be demonstrated. These clusters of arguments make the association between psychiatric diseases and aPL increasingly plausible. However, a considerable amount of clinical research must still be performed in accordance with the highest standards of methodological quality. The therapeutic management of this association, in terms of both prevention and cure, currently remains unresolved.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1788696

    Thrombotic events striking the central nervous system are clinical criteria for the antiphospholipid syndrome (APS). Besides these, neuropsychiatric non-APS criteria manifestations are increasingly described in patients with persistently positive antiphospholipid antibodies (aPL). Among these are psychiatric manifestations. Animal models mainly describe hyperactive behavior and anxiety associated with hippocampal abnormalities. Cases of associations with psychosis, mood disorders, bipolarity, anxiety, obsessive–compulsive behavior, and depression have been reported but are still rare. Systematic human clinical association studies are concordant with a risk of psychosis, depression (simple to major), and anxiety disorders, but these are limited and of inconstant methodological quality. Brain imaging in patients, also insufficiently investigated, shows early signs of hypoperfusion and of subtle diffuse white matter changes compatible with an alteration of the axonal structure and changes in the myelin sheath. Direct interactions of aPL with the brain cells, both on cell lines and on animal and human brain biopsies, targeting both glial cells, astrocytes, and neurons, can be demonstrated. These clusters of arguments make the association between psychiatric diseases and aPL increasingly plausible. However, a considerable amount of clinical research must still be performed in accordance with the highest standards of methodological quality. The therapeutic management of this association, in terms of both prevention and cure, currently remains unresolved.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Antiphospholipid Antibodies in Mental DisordersGris, Jean-ChristopheChéa, MathiasBouvier, SylviePereira, Fabricio R.DOI:10.1055/s-0044-1788696Semin Thromb Hemost ; : -2024-07-24T20:09:01+01:00Seminars in Thrombosis and Hemostasis2024-07-24T20:09:01+01:00eFirst
    Review Article
    10.1055/s-0044-1788696http://dx.doi.org/10.1055/s-0044-1788696
    New STH 2023 Impact Factor, Most Highly Cited Papers, and Other Journal Metricshttp://dx.doi.org/10.1055/s-0044-1788566Semin Thromb Hemost
    DOI: 10.1055/s-0044-1788566



    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Full text

    ]]>
    New STH 2023 Impact Factor, Most Highly Cited Papers, and Other Journal MetricsFavaloro, Emmanuel J.DOI:10.1055/s-0044-1788566Semin Thromb Hemost ; : -2024-07-19T18:38:12+01:00Seminars in Thrombosis and Hemostasis2024-07-19T18:38:12+01:00eFirst
    Editorial
    10.1055/s-0044-1788566http://dx.doi.org/10.1055/s-0044-1788566
    Mendelian Randomization Provides No Evidence for the Bidirectional Relationship between Type 2 Diabetes and Venous Thromboembolism in East Asians and African Americanshttp://dx.doi.org/10.1055/s-0044-1788568Semin Thromb Hemost
    DOI: 10.1055/s-0044-1788568



    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Full text

    ]]>
    Mendelian Randomization Provides No Evidence for the Bidirectional Relationship between Type 2 Diabetes and Venous Thromboembolism in East Asians and African AmericansLu, JiawenWang, ZhenqianDOI:10.1055/s-0044-1788568Semin Thromb Hemost ; : -2024-07-19T18:38:11+01:00Seminars in Thrombosis and Hemostasis2024-07-19T18:38:11+01:00eFirst
    Letter to the Editor
    10.1055/s-0044-1788568http://dx.doi.org/10.1055/s-0044-1788568
    Laboratory Diagnostics for Thrombosis and Hemostasis Testing—Part 3http://dx.doi.org/10.1055/s-0044-1788567Semin Thromb Hemost
    DOI: 10.1055/s-0044-1788567



    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Full text

    ]]>
    Laboratory Diagnostics for Thrombosis and Hemostasis Testing—Part 3Smock, Kristi J.Moffat, Karen A.DOI:10.1055/s-0044-1788567Semin Thromb Hemost ; : -2024-07-19T18:38:13+01:00Seminars in Thrombosis and Hemostasis2024-07-19T18:38:13+01:00eFirst
    Preface
    10.1055/s-0044-1788567http://dx.doi.org/10.1055/s-0044-1788567
    The Impact of Emotional Responses on Female Reproduction: Fibrinolysis in the Spotlighthttp://dx.doi.org/10.1055/s-0044-1788324Fibrinolytic enzymes modify various substrates required for tissue remodeling, playing a crucial role in mechanisms underlying resilience, reward processing, ovulation, embryo implantation, and placentation. Individuals with low resilience and reduced reward responsiveness, when exposed to chronic stress, are at increased risk of experiencing a range of negative emotions. Chronic anxiety and melancholia are examples of negative emotions associated with hypercortisolism, while fear and atypical depression are characterized by systemic inflammation. Both cortisol and inflammatory cytokines stimulate the production of plasminogen activator inhibitor-1 (PAI-1), a potent fibrinolysis inhibitor. Chronic anxiety, fear, and depression are among the many hypofibrinolytic conditions increasing the risk of oligo-anovulation, miscarriage, fetal growth restriction, and preeclampsia. Although significant, the impact of negative emotions on implantation is not as obvious as on ovulation or placentation. Other hypofibrinolytic conditions that may affect female reproduction through mechanisms dependent or independent of PAI-1 include metabolic disturbances (e.g., due to consumption of highly palatable foods, often used to alleviate negative affect), inflammation, hyperhomocysteinemia, hypothyroidism, hypercortisolism, antiphospholipid antibodies, and the 4G allele of the PAI-1 gene. Benzodiazepines and antidepressants should be used with caution in the first trimester as this combination may cause malformations. Also, selective serotonin reuptake inhibitors have fibrinolytic properties that increase the risk of bleeding after surgical procedures. Psychological interventions, especially group therapy, are effective in the prevention of reproductive disorders. Controlled trials are needed to test the hypothesis that female reproductive health depends on psychological well-being, a balanced diet and physical activity, suppression of inflammation and autoantibodies, and homocysteine and hormonal homeostasis.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1788324

    Fibrinolytic enzymes modify various substrates required for tissue remodeling, playing a crucial role in mechanisms underlying resilience, reward processing, ovulation, embryo implantation, and placentation. Individuals with low resilience and reduced reward responsiveness, when exposed to chronic stress, are at increased risk of experiencing a range of negative emotions. Chronic anxiety and melancholia are examples of negative emotions associated with hypercortisolism, while fear and atypical depression are characterized by systemic inflammation. Both cortisol and inflammatory cytokines stimulate the production of plasminogen activator inhibitor-1 (PAI-1), a potent fibrinolysis inhibitor. Chronic anxiety, fear, and depression are among the many hypofibrinolytic conditions increasing the risk of oligo-anovulation, miscarriage, fetal growth restriction, and preeclampsia. Although significant, the impact of negative emotions on implantation is not as obvious as on ovulation or placentation. Other hypofibrinolytic conditions that may affect female reproduction through mechanisms dependent or independent of PAI-1 include metabolic disturbances (e.g., due to consumption of highly palatable foods, often used to alleviate negative affect), inflammation, hyperhomocysteinemia, hypothyroidism, hypercortisolism, antiphospholipid antibodies, and the 4G allele of the PAI-1 gene. Benzodiazepines and antidepressants should be used with caution in the first trimester as this combination may cause malformations. Also, selective serotonin reuptake inhibitors have fibrinolytic properties that increase the risk of bleeding after surgical procedures. Psychological interventions, especially group therapy, are effective in the prevention of reproductive disorders. Controlled trials are needed to test the hypothesis that female reproductive health depends on psychological well-being, a balanced diet and physical activity, suppression of inflammation and autoantibodies, and homocysteine and hormonal homeostasis.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    The Impact of Emotional Responses on Female Reproduction: Fibrinolysis in the SpotlightHoirisch-Clapauch, SilviaDOI:10.1055/s-0044-1788324Semin Thromb Hemost ; : -2024-07-19T18:38:12+01:00Seminars in Thrombosis and Hemostasis2024-07-19T18:38:12+01:00eFirst
    Review Article
    10.1055/s-0044-1788324http://dx.doi.org/10.1055/s-0044-1788324
    2023 Eberhard F. Mammen Award Announcements: Part II – Young Investigator Awardshttp://dx.doi.org/10.1055/s-0044-1787989Semin Thromb Hemost
    DOI: 10.1055/s-0044-1787989



    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Full text

    ]]>
    2023 Eberhard F. Mammen Award Announcements: Part II – Young Investigator AwardsFavaloro, Emmanuel J.DOI:10.1055/s-0044-1787989Semin Thromb Hemost ; : -2024-07-01T06:34:19+01:00Seminars in Thrombosis and Hemostasis2024-07-01T06:34:19+01:00eFirst
    Editorial
    10.1055/s-0044-1787989http://dx.doi.org/10.1055/s-0044-1787989
    Point-of-Care Testing in Patients with Hereditary Disorders of Primary Hemostasis: A Narrative Reviewhttp://dx.doi.org/10.1055/s-0044-1787976Inherited disorders of primary hemostasis, such as von Willebrand disease and congenital platelet disorders, can cause extensive, typically mucocutaneous bleeding. Assays to diagnose and monitor these disorders, such as von Willebrand factor activity assays and light transmission aggregometry, are performed in specialized hemostasis laboratories but are commonly not available in local hospitals. Due to the complexity and relative scarcity of these conventional assays, point-of-care tests (POCT) might be an attractive alternative in patients with hereditary bleeding disorders. POCTs, such as thromboelastography, are increasingly used to assess hemostasis in patients with acquired hemostatic defects, aiding clinical decision-making in critical situations, such as during surgery or childbirth. In comparison, the use of these assays in patients with hereditary hemostasis defects remains relatively unexplored. This review aims to give an overview of point-of-care hemostasis tests in patients with hereditary disorders of primary hemostasis. A summary of the literature reporting on the performance of currently available and experimental POCTs in these disorders is given, and the potential utility of the assays in various use scenarios is discussed. Altogether, the studies included in this review reveal that several POCTs are capable of identifying and monitoring severe defects in the primary hemostasis, while a POCT that can reliably detect milder defects of primary hemostasis is currently lacking. A better understanding of the strengths and limitations of POCTs in assessing hereditary defects of primary hemostasis is needed, after which these tests may become available for clinical practice, potentially targeting a large group of patients with milder defects of primary hemostasis.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1787976

    Inherited disorders of primary hemostasis, such as von Willebrand disease and congenital platelet disorders, can cause extensive, typically mucocutaneous bleeding. Assays to diagnose and monitor these disorders, such as von Willebrand factor activity assays and light transmission aggregometry, are performed in specialized hemostasis laboratories but are commonly not available in local hospitals. Due to the complexity and relative scarcity of these conventional assays, point-of-care tests (POCT) might be an attractive alternative in patients with hereditary bleeding disorders. POCTs, such as thromboelastography, are increasingly used to assess hemostasis in patients with acquired hemostatic defects, aiding clinical decision-making in critical situations, such as during surgery or childbirth. In comparison, the use of these assays in patients with hereditary hemostasis defects remains relatively unexplored. This review aims to give an overview of point-of-care hemostasis tests in patients with hereditary disorders of primary hemostasis. A summary of the literature reporting on the performance of currently available and experimental POCTs in these disorders is given, and the potential utility of the assays in various use scenarios is discussed. Altogether, the studies included in this review reveal that several POCTs are capable of identifying and monitoring severe defects in the primary hemostasis, while a POCT that can reliably detect milder defects of primary hemostasis is currently lacking. A better understanding of the strengths and limitations of POCTs in assessing hereditary defects of primary hemostasis is needed, after which these tests may become available for clinical practice, potentially targeting a large group of patients with milder defects of primary hemostasis.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Point-of-Care Testing in Patients with Hereditary Disorders of Primary Hemostasis: A Narrative ReviewBavinck, Aernoud P.Heerde, Waander vanSchols, Saskia E.M.DOI:10.1055/s-0044-1787976Semin Thromb Hemost ; : -2024-07-01T06:34:21+01:00Seminars in Thrombosis and Hemostasis2024-07-01T06:34:21+01:00eFirst
    Review Article
    10.1055/s-0044-1787976http://dx.doi.org/10.1055/s-0044-1787976
    Current Diagnosis of von Willebrand Disease in Italy: 3 Years Following the Release of the International Guidelineshttp://dx.doi.org/10.1055/s-0044-1787841The American Society of Hematology–International Society on Thrombosis and Haemostasis–National Hemophilia Foundation–World Federation of Hemophilia 2021 International Guidelines (IGL) on von Willebrand disease (VWD) have pointed out many challenges, mainly in the diagnostic approach of VWD patients. To determine the impact of these IGL on the current clinical and laboratory diagnosis of Italian VWD patients, we have recently conducted a survey among 43 centers affiliated with the Italian Association of Hemophilia Centers (AICE). Directors and colleagues responsible for the management of VWD patients were invited to report in a detailed questionnaire how IGL recommendations about the assessment of the specific activities of von Willebrand Factor (VWF) could be applied at their local sites. Results from such a survey showed that bleeding assessment tools, VWF antigen, and factor VIII procoagulant are currently in use in all centers. The automated assays for platelet-dependent VWF activity with or without ristocetin described in IGL have been used since 2021 in 37/43 (86%) centers. Among other laboratory tests, VWF collagen binding, ristocetin-induced platelet agglutination, multimeric analysis, VWF propeptide, VWF:FVIII binding assay were available in 49, 63, 26, 7, and 28% of AICE, respectively. Analyses of VWF gene defects are available only at 3/43 (7%) centers. Desmopressin (DDAVP) infusion trials at diagnosis, with measurements of VWF activities at 1 and 4 hours post-DDAVP, is currently performed at 38/43 (88%) centers. Based on this information, a simplified clinical diagnosis using a few automated tests before and after DDAVP has been proposed. Such a diagnostic approach will be validated prospectively in a large cohort of Italian VWD patients.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1787841

    The American Society of Hematology–International Society on Thrombosis and Haemostasis–National Hemophilia Foundation–World Federation of Hemophilia 2021 International Guidelines (IGL) on von Willebrand disease (VWD) have pointed out many challenges, mainly in the diagnostic approach of VWD patients. To determine the impact of these IGL on the current clinical and laboratory diagnosis of Italian VWD patients, we have recently conducted a survey among 43 centers affiliated with the Italian Association of Hemophilia Centers (AICE). Directors and colleagues responsible for the management of VWD patients were invited to report in a detailed questionnaire how IGL recommendations about the assessment of the specific activities of von Willebrand Factor (VWF) could be applied at their local sites. Results from such a survey showed that bleeding assessment tools, VWF antigen, and factor VIII procoagulant are currently in use in all centers. The automated assays for platelet-dependent VWF activity with or without ristocetin described in IGL have been used since 2021 in 37/43 (86%) centers. Among other laboratory tests, VWF collagen binding, ristocetin-induced platelet agglutination, multimeric analysis, VWF propeptide, VWF:FVIII binding assay were available in 49, 63, 26, 7, and 28% of AICE, respectively. Analyses of VWF gene defects are available only at 3/43 (7%) centers. Desmopressin (DDAVP) infusion trials at diagnosis, with measurements of VWF activities at 1 and 4 hours post-DDAVP, is currently performed at 38/43 (88%) centers. Based on this information, a simplified clinical diagnosis using a few automated tests before and after DDAVP has been proposed. Such a diagnostic approach will be validated prospectively in a large cohort of Italian VWD patients.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Current Diagnosis of von Willebrand Disease in Italy: 3 Years Following the Release of the International GuidelinesFederici, Augusto B.DOI:10.1055/s-0044-1787841Semin Thromb Hemost ; : -2024-07-01T06:34:20+01:00Seminars in Thrombosis and Hemostasis2024-07-01T06:34:20+01:00eFirst
    Review Article
    10.1055/s-0044-1787841http://dx.doi.org/10.1055/s-0044-1787841
    Pharmacotherapy for Venous Thromboprophylaxis following Total Hip or Knee Arthroplasty: A Systematic Review and Network Meta-analysishttp://dx.doi.org/10.1055/s-0044-1787996The optimal pharmacological prophylaxis for venous thromboembolism (VTE) after hip or knee arthroplasty is uncertain. We conducted a systematic review and network meta-analysis to compare the efficacy and safety of various medications. We searched multiple databases for randomized clinical trials (RCTs) comparing medications (including factor Xa inhibitors, factor IIa inhibitor, warfarin, unfractionated heparin [UFH], low-molecular-weight heparin [LMWH], aspirin, pentasaccharide) for VTE prophylaxis post-arthroplasty. Outcomes included any postoperative VTE identified with screening, major bleeding, and death. We used LMWH as the main comparator for analysis and performed trial sequential analysis (TSA) for each pairwise comparison. Certainty of evidence was assessed using GRADE (Grading of Recommendations, Assessments, Developments and Evaluations). We analyzed 70 RCTs (55,841 participants). Factor Xa inhibitors decreased postoperative VTE significantly compared with LMWH (odds ratio [OR]: 0.55, 95% confidence interval [CI]: 0.44–0.68, high certainty). Pentasaccharides probably reduce VTE (OR: 0.61, 95% CI: 0.36–1.02, moderate certainty), while the factor IIa inhibitor dabigatran may reduce VTE (OR: 0.75, 95% CI: 0.40–1.42, low certainty). UFH probably increases VTE compared with LMWH (OR: 1.31, 95% CI: 0.91–1.89, moderate certainty), and other agents like warfarin, aspirin, placebo, and usual care without thromboprophylaxis increase VTE (high certainty). Factor Xa inhibitors may not significantly affect major bleeding compared with LMWH (OR: 1.06, 95% CI: 0.81–1.39, low certainty). No medications had a notable effect on mortality compared with LMWH (very low certainty). TSA suggests sufficient evidence for the benefit of factor Xa inhibitors over LMWH for VTE prevention. Compared with LMWH and aspirin, factor Xa inhibitors are associated with reduced VTE after hip or knee arthroplasty, without an increase in bleeding and likely no impact on mortality.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1787996

    The optimal pharmacological prophylaxis for venous thromboembolism (VTE) after hip or knee arthroplasty is uncertain. We conducted a systematic review and network meta-analysis to compare the efficacy and safety of various medications. We searched multiple databases for randomized clinical trials (RCTs) comparing medications (including factor Xa inhibitors, factor IIa inhibitor, warfarin, unfractionated heparin [UFH], low-molecular-weight heparin [LMWH], aspirin, pentasaccharide) for VTE prophylaxis post-arthroplasty. Outcomes included any postoperative VTE identified with screening, major bleeding, and death. We used LMWH as the main comparator for analysis and performed trial sequential analysis (TSA) for each pairwise comparison. Certainty of evidence was assessed using GRADE (Grading of Recommendations, Assessments, Developments and Evaluations). We analyzed 70 RCTs (55,841 participants). Factor Xa inhibitors decreased postoperative VTE significantly compared with LMWH (odds ratio [OR]: 0.55, 95% confidence interval [CI]: 0.44–0.68, high certainty). Pentasaccharides probably reduce VTE (OR: 0.61, 95% CI: 0.36–1.02, moderate certainty), while the factor IIa inhibitor dabigatran may reduce VTE (OR: 0.75, 95% CI: 0.40–1.42, low certainty). UFH probably increases VTE compared with LMWH (OR: 1.31, 95% CI: 0.91–1.89, moderate certainty), and other agents like warfarin, aspirin, placebo, and usual care without thromboprophylaxis increase VTE (high certainty). Factor Xa inhibitors may not significantly affect major bleeding compared with LMWH (OR: 1.06, 95% CI: 0.81–1.39, low certainty). No medications had a notable effect on mortality compared with LMWH (very low certainty). TSA suggests sufficient evidence for the benefit of factor Xa inhibitors over LMWH for VTE prevention. Compared with LMWH and aspirin, factor Xa inhibitors are associated with reduced VTE after hip or knee arthroplasty, without an increase in bleeding and likely no impact on mortality.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Pharmacotherapy for Venous Thromboprophylaxis following Total Hip or Knee Arthroplasty: A Systematic Review and Network Meta-analysisYong, Bryan Song JunLing, Ryan RuiyangLi, RuiqiPoh, Jane WenjinTan, Chuen SengHo, Sean Wei LoongRochwerg, BramArya, RoopenRamanathan, KollengodeFan, Bingwen EugeneDOI:10.1055/s-0044-1787996Semin Thromb Hemost ; : -2024-07-01T06:34:20+01:00Seminars in Thrombosis and Hemostasis2024-07-01T06:34:20+01:00eFirst
    Review Article
    10.1055/s-0044-1787996http://dx.doi.org/10.1055/s-0044-1787996
    Emicizumab in Type 3 von Willebrand Disease: Report of a Case with an Alloantibody and Literature Reviewhttp://dx.doi.org/10.1055/s-0044-1787662Type 3 von Willebrand disease (VWD), the most severe form of VWD, is an inherited recessive bleeding disorder caused by the complete deficiency of von Willebrand factor (VWF). The reported prevalence is 1 per million but varies worldwide according to the frequency of consanguineous marriages. The clinical phenotype is characterized not only by mucocutaneous bleedings, but also by hemarthroses and muscle hematoma, as in patients with moderate hemophilia. Long-term prophylaxis with factor (F)VIII/VWF concentrates is recommended in patients with a history of severe and frequent bleeds. A rare complication of replacement therapy is the development of alloantibodies against VWF, with the consequences of an ineffective therapy and risk of anaphylactic reactions upon treatment. Emicizumab is the first bispecific monoclonal antibody that mimics FVIII coagulant activity and is approved for prophylaxis of bleeding in patients with inherited hemophilia A with or without inhibitors and recently also for acquired hemophilia. In this manuscript we report and discuss available data in the literature on the use of emicizumab in type 3 VWD and describe the case of a female patient with type 3 VWD with a history of alloantibodies against VWF and posttransfusion anaphylaxis, recently and successfully put on off-label prophylaxis with emicizumab.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1787662

    Type 3 von Willebrand disease (VWD), the most severe form of VWD, is an inherited recessive bleeding disorder caused by the complete deficiency of von Willebrand factor (VWF). The reported prevalence is 1 per million but varies worldwide according to the frequency of consanguineous marriages. The clinical phenotype is characterized not only by mucocutaneous bleedings, but also by hemarthroses and muscle hematoma, as in patients with moderate hemophilia. Long-term prophylaxis with factor (F)VIII/VWF concentrates is recommended in patients with a history of severe and frequent bleeds. A rare complication of replacement therapy is the development of alloantibodies against VWF, with the consequences of an ineffective therapy and risk of anaphylactic reactions upon treatment. Emicizumab is the first bispecific monoclonal antibody that mimics FVIII coagulant activity and is approved for prophylaxis of bleeding in patients with inherited hemophilia A with or without inhibitors and recently also for acquired hemophilia. In this manuscript we report and discuss available data in the literature on the use of emicizumab in type 3 VWD and describe the case of a female patient with type 3 VWD with a history of alloantibodies against VWF and posttransfusion anaphylaxis, recently and successfully put on off-label prophylaxis with emicizumab.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Emicizumab in Type 3 von Willebrand Disease: Report of a Case with an Alloantibody and Literature ReviewGiuffrida, Anna C.Siboni, Simona M.Baronciani, LucianoPoli, GiovanniGandini, GiorgioPeyvandi, FloraDOI:10.1055/s-0044-1787662Semin Thromb Hemost ; : -2024-06-27T12:06:50+01:00Seminars in Thrombosis and Hemostasis2024-06-27T12:06:50+01:00eFirst
    Review Article
    10.1055/s-0044-1787662http://dx.doi.org/10.1055/s-0044-1787662
    Stress-Induced Hypercoagulability: Insights from Epidemiological and Mechanistic Studies, and Clinical Integrationhttp://dx.doi.org/10.1055/s-0044-1787660By integrating findings from comprehensive reviews, meta-analyses, and cutting-edge genetic studies, this article illuminates the significance of stress-induced hypercoagulability in clinical medicine. In particular, the findings from numerous prospective cohort studies indicate that stress and hemostatic factors of a hypercoagulable state are associated with increased incident risk and poor prognosis for atherosclerotic cardiovascular disease and venous thromboembolism. Mendelian randomization studies suggest that these associations are partially causal. The review synthesizes extensive research on the link between acute and chronic stress and hypercoagulability, outlining a potential pathway from stress to thrombosis risk. Consistent with the allostatic load concept, acute stress-induced hypercoagulability, initially adaptive, can turn maladaptive under chronic stress or excessive acute stress, leading to arterial or venous thrombotic events. Individuals with predisposing factors, including atherosclerosis, thrombophilia, or immobilization, may exhibit an increased risk of thrombotic disease during stress. Contextual sociodemographic characteristics, the stress experience, and coping resources additionally modulate the extent of stress-induced hypercoagulability. Research into the neuroendocrine, cellular, and molecular bases reveals how stress influences platelet activation coagulation and fibrinolysis. The activation of the sympathetic nervous system and the hypothalamic–pituitary–adrenal axis, along with vagal withdrawal, and the effects of catecholamines, cortisol, and vasopressin, are the central mechanisms involved. Hemoconcentration, inflammation, endothelial dysfunction, and thrombopoiesis additionally contribute to stress-induced hypercoagulability. Further research is needed to prove a causal link between chronic stress and hypercoagulability. This includes exploring its implications for the prevention and management of thrombotic diseases in stressed individuals, with a focus on developing effective psychosocial and pharmacological interventions.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1787660

    By integrating findings from comprehensive reviews, meta-analyses, and cutting-edge genetic studies, this article illuminates the significance of stress-induced hypercoagulability in clinical medicine. In particular, the findings from numerous prospective cohort studies indicate that stress and hemostatic factors of a hypercoagulable state are associated with increased incident risk and poor prognosis for atherosclerotic cardiovascular disease and venous thromboembolism. Mendelian randomization studies suggest that these associations are partially causal. The review synthesizes extensive research on the link between acute and chronic stress and hypercoagulability, outlining a potential pathway from stress to thrombosis risk. Consistent with the allostatic load concept, acute stress-induced hypercoagulability, initially adaptive, can turn maladaptive under chronic stress or excessive acute stress, leading to arterial or venous thrombotic events. Individuals with predisposing factors, including atherosclerosis, thrombophilia, or immobilization, may exhibit an increased risk of thrombotic disease during stress. Contextual sociodemographic characteristics, the stress experience, and coping resources additionally modulate the extent of stress-induced hypercoagulability. Research into the neuroendocrine, cellular, and molecular bases reveals how stress influences platelet activation coagulation and fibrinolysis. The activation of the sympathetic nervous system and the hypothalamic–pituitary–adrenal axis, along with vagal withdrawal, and the effects of catecholamines, cortisol, and vasopressin, are the central mechanisms involved. Hemoconcentration, inflammation, endothelial dysfunction, and thrombopoiesis additionally contribute to stress-induced hypercoagulability. Further research is needed to prove a causal link between chronic stress and hypercoagulability. This includes exploring its implications for the prevention and management of thrombotic diseases in stressed individuals, with a focus on developing effective psychosocial and pharmacological interventions.
    [...]

    Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Stress-Induced Hypercoagulability: Insights from Epidemiological and Mechanistic Studies, and Clinical Integrationvon Känel, RolandDOI:10.1055/s-0044-1787660Semin Thromb Hemost ; : -2024-06-24T07:08:32+01:00Seminars in Thrombosis and Hemostasis2024-06-24T07:08:32+01:00eFirst
    Review Article
    10.1055/s-0044-1787660http://dx.doi.org/10.1055/s-0044-1787660
    Hemostasis Testing in the Emergency Department: A Narrative Reviewhttp://dx.doi.org/10.1055/s-0044-1787661Routine laboratory screening is typically performed at initial evaluation of the vast majority of presentations to the emergency department (ED). These laboratory results are crucial to the diagnostic process, as they may influence up to 70% of clinical decisions. However, despite the usefulness of biological assessments, many tests performed are inappropriate or of doubtful clinical relevance. This overutilization rate of laboratory testing in hospitals, which represents a significant medical–economic burden, ranges from 20 to 67%, with coagulation tests at the top of the list. While reviews frequently focus on nonintensive care units, there are few published assessments of emergency-specific interventions or guidelines/guidance to date. The aim of this review is to highlight current recommendations for hemostasis evaluation in the emergency setting with a specific analysis of common situations leading to ED admissions, such as suspected venous thrombosis or severe bleeding. We revisit the evidence related to the assessment of patient's hemostatic capacity based on comprehensive history taking and physical examination as well as best practice recommendations for blood sample collection to ensure the reliability of results. This review also includes an examination of various currently available point of care tests and a comprehensive discussion on indications, limitations, and interpretation of these tests.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1787661

    Routine laboratory screening is typically performed at initial evaluation of the vast majority of presentations to the emergency department (ED). These laboratory results are crucial to the diagnostic process, as they may influence up to 70% of clinical decisions. However, despite the usefulness of biological assessments, many tests performed are inappropriate or of doubtful clinical relevance. This overutilization rate of laboratory testing in hospitals, which represents a significant medical–economic burden, ranges from 20 to 67%, with coagulation tests at the top of the list. While reviews frequently focus on nonintensive care units, there are few published assessments of emergency-specific interventions or guidelines/guidance to date. The aim of this review is to highlight current recommendations for hemostasis evaluation in the emergency setting with a specific analysis of common situations leading to ED admissions, such as suspected venous thrombosis or severe bleeding. We revisit the evidence related to the assessment of patient's hemostatic capacity based on comprehensive history taking and physical examination as well as best practice recommendations for blood sample collection to ensure the reliability of results. This review also includes an examination of various currently available point of care tests and a comprehensive discussion on indications, limitations, and interpretation of these tests.
    [...]

    Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Hemostasis Testing in the Emergency Department: A Narrative ReviewThonon, HenriVan Nieuwenhove, MichaelThachil, JeckoLippi, GiuseppeHardy, MichaelMullier, FrançoisDOI:10.1055/s-0044-1787661Semin Thromb Hemost ; : -2024-06-19T13:43:18+01:00Seminars in Thrombosis and Hemostasis2024-06-19T13:43:18+01:00eFirst
    Review Article
    10.1055/s-0044-1787661http://dx.doi.org/10.1055/s-0044-1787661
    Dosing Intensity of Anticoagulants for the Prevention and Treatment of Venous Thromboembolism and the Prevention of Stroke in Atrial Fibrillation: Why is There a Difference?http://dx.doi.org/10.1055/s-0044-1787726Semin Thromb Hemost
    DOI: 10.1055/s-0044-1787726



    Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

    Article in Thieme eJournals:
    Table of contents  |  Full text

    ]]>
    Dosing Intensity of Anticoagulants for the Prevention and Treatment of Venous Thromboembolism and the Prevention of Stroke in Atrial Fibrillation: Why is There a Difference?Hirsh, JackGodoy, AlejandroChan, Noel C.DOI:10.1055/s-0044-1787726Semin Thromb Hemost ; : -2024-06-18T13:34:44+01:00Seminars in Thrombosis and Hemostasis2024-06-18T13:34:44+01:00eFirst
    Commentary
    10.1055/s-0044-1787726http://dx.doi.org/10.1055/s-0044-1787726
    Platelet Pathophysiology: Unexpected New Research Directionshttp://dx.doi.org/10.1055/s-0044-1787663Semin Thromb Hemost
    DOI: 10.1055/s-0044-1787663



    Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

    Article in Thieme eJournals:
    Table of contents  |  Full text

    ]]>
    Platelet Pathophysiology: Unexpected New Research DirectionsMichelson, Alan D.Frelinger III, Andrew L.Haynes, Robin L.Kinney, Hannah C.Gremmel, ThomasDOI:10.1055/s-0044-1787663Semin Thromb Hemost ; : -2024-06-18T13:34:45+01:00Seminars in Thrombosis and Hemostasis2024-06-18T13:34:45+01:00eFirst
    Commentary
    10.1055/s-0044-1787663http://dx.doi.org/10.1055/s-0044-1787663
    Patient Perspective on Disease Burden and Gene Therapy for Hemophilia A and B: The “Haemvolution for Patients” Italian Surveyhttp://dx.doi.org/10.1055/s-0044-1787664Hemophilia is a rare X-linked congenital bleeding disorder due to a deficiency of factor VIII (hemophilia A [HA]) or factor IX (hemophilia B [HB]). Replacement and nonreplacement treatments are available but have limitations. Gene therapy (GT) provides an effective, long-term, single-dose treatment option, now approaching clinical practice. This study aimed to understand patient perspectives on GT for HA and HB in Italy using a qualitative questionnaire distributed through Italian patient associations, addressing patient views on daily life, treatments, unmet needs, quality of life (QoL), and GT for hemophilia. In total, 141 participants had HA, and 14 had HB (severe 78.6%). Daily life was most affected by pain and/or joint function limitations (57.5% of participants), high infusion frequency (42.5%), management of breakthrough bleeding episodes (40.3%), and anxiety/fear of severe or sudden bleeding (38.8%). Despite current treatments, about half of the participants experienced three or more annual bleeding episodes. Most participants knew of GT (87.2%) and expected improvements in QoL (60.5%), reduced frequency of current treatments (53.5%), and a permanent cure (49.1%); 46.4% were unaware of its once-off dosage and 46.4% were not concerned about the costs they anticipated to be associated with GT. Although several fears were reported, 25.0% of participants were willing to undergo GT with the support of a multidisciplinary team. This survey provided valuable insight into patient perspectives on hemophilia and GT in Italy. Overall, relevant proportions of patients still experience limitations affecting their daily life. Most were positive about GT and anticipated improvements in their clinical outcomes and QoL.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1787664

    Hemophilia is a rare X-linked congenital bleeding disorder due to a deficiency of factor VIII (hemophilia A [HA]) or factor IX (hemophilia B [HB]). Replacement and nonreplacement treatments are available but have limitations. Gene therapy (GT) provides an effective, long-term, single-dose treatment option, now approaching clinical practice. This study aimed to understand patient perspectives on GT for HA and HB in Italy using a qualitative questionnaire distributed through Italian patient associations, addressing patient views on daily life, treatments, unmet needs, quality of life (QoL), and GT for hemophilia. In total, 141 participants had HA, and 14 had HB (severe 78.6%). Daily life was most affected by pain and/or joint function limitations (57.5% of participants), high infusion frequency (42.5%), management of breakthrough bleeding episodes (40.3%), and anxiety/fear of severe or sudden bleeding (38.8%). Despite current treatments, about half of the participants experienced three or more annual bleeding episodes. Most participants knew of GT (87.2%) and expected improvements in QoL (60.5%), reduced frequency of current treatments (53.5%), and a permanent cure (49.1%); 46.4% were unaware of its once-off dosage and 46.4% were not concerned about the costs they anticipated to be associated with GT. Although several fears were reported, 25.0% of participants were willing to undergo GT with the support of a multidisciplinary team. This survey provided valuable insight into patient perspectives on hemophilia and GT in Italy. Overall, relevant proportions of patients still experience limitations affecting their daily life. Most were positive about GT and anticipated improvements in their clinical outcomes and QoL.
    [...]

    Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Patient Perspective on Disease Burden and Gene Therapy for Hemophilia A and B: The “Haemvolution for Patients” Italian SurveyMansueto, Maria FrancescaBigi, SarahFollino, MarcoLupi, AngeloCoppola, AntonioDOI:10.1055/s-0044-1787664Semin Thromb Hemost ; : -2024-06-18T13:34:45+01:00Seminars in Thrombosis and Hemostasis2024-06-18T13:34:45+01:00eFirst
    Review Article
    10.1055/s-0044-1787664http://dx.doi.org/10.1055/s-0044-1787664
    Laboratory Testing for Fibrinogen Disorders: From Routine Investigations to Research Studieshttp://dx.doi.org/10.1055/s-0044-1787725Congenital and acquired fibrinogen disorders often have heterogeneous clinical phenotypes and are challenging from a laboratory perspective. Fibrinogen determination using the Clauss method remains the gold standard, while the reproducibility and significance of the thrombin time and the reptilase time are limited. Molecular testing for causative mutations in fibrinogen genes is now recommended to confirm the diagnosis of congenital fibrinogen disorders. Research assays are used to evaluate alterations to fibrin formation and properties of plasma and purified fibrinogen-derived clots, characterized by fiber thickness, the number of branches, and pore sizes. Fibrin clot permeability (permeation, porosity) using a hydrostatic pressure system represents the most commonly used method for evaluating fibrin network density. Reduced clot permeability, which denotes the reduced size of an average pore in the network, results in tighter fibrin networks, typically associated with impaired susceptibility to lysis, leading to a thrombotic tendency. Biophysical properties of fibrin clots are largely assessed using rheometry, with atomic force microscopy and nanorheology being increasingly used in disease states. Thromboelastography and thromboelastometry, a simple modification of rheometry, have been used, mainly in intensive care units, for more than 50 years. Given growing evidence for altered fibrin clot properties in diseases with elevated risk of venous and arterial thromboembolism and in some bleeding disorders, further work on standardization and validation of the assessment of fibrin clot characteristics is needed. This review summarizes the current methods used to evaluate fibrinogen abnormalities in both diagnostic and research laboratories.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1787725

    Congenital and acquired fibrinogen disorders often have heterogeneous clinical phenotypes and are challenging from a laboratory perspective. Fibrinogen determination using the Clauss method remains the gold standard, while the reproducibility and significance of the thrombin time and the reptilase time are limited. Molecular testing for causative mutations in fibrinogen genes is now recommended to confirm the diagnosis of congenital fibrinogen disorders. Research assays are used to evaluate alterations to fibrin formation and properties of plasma and purified fibrinogen-derived clots, characterized by fiber thickness, the number of branches, and pore sizes. Fibrin clot permeability (permeation, porosity) using a hydrostatic pressure system represents the most commonly used method for evaluating fibrin network density. Reduced clot permeability, which denotes the reduced size of an average pore in the network, results in tighter fibrin networks, typically associated with impaired susceptibility to lysis, leading to a thrombotic tendency. Biophysical properties of fibrin clots are largely assessed using rheometry, with atomic force microscopy and nanorheology being increasingly used in disease states. Thromboelastography and thromboelastometry, a simple modification of rheometry, have been used, mainly in intensive care units, for more than 50 years. Given growing evidence for altered fibrin clot properties in diseases with elevated risk of venous and arterial thromboembolism and in some bleeding disorders, further work on standardization and validation of the assessment of fibrin clot characteristics is needed. This review summarizes the current methods used to evaluate fibrinogen abnormalities in both diagnostic and research laboratories.
    [...]

    Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Laboratory Testing for Fibrinogen Disorders: From Routine Investigations to Research StudiesUndas, AnettaDOI:10.1055/s-0044-1787725Semin Thromb Hemost ; : -2024-06-18T13:34:46+01:00Seminars in Thrombosis and Hemostasis2024-06-18T13:34:46+01:00eFirst
    Review Article
    10.1055/s-0044-1787725http://dx.doi.org/10.1055/s-0044-1787725
    Diagnosis and Treatment of Autoimmune Acquired Coagulation Factor Deficiencies: An Evidence-Based Review of Japanese Practicehttp://dx.doi.org/10.1055/s-0044-1787188Among the acquired coagulation factor deficiencies, autoimmune coagulation factor deficiencies (AiCFD) are rare and result from autoantibody production against coagulation factors. In Japan, a nationwide survey on AiCFD has been conducted since 2009. Autoimmune factor XIII, factor VIII, von Willebrand factor, factor V, and factor X deficiencies (AiF13D, AiF8D, AiVWFD, AiF5D, and AiF10D, respectively) have been enacted as “designated intractable disease-282.” The incidence of AiF8D, AiF13D, and AiF5D was 1.83, 0.044, and 0.038 per million people/year, respectively, whereas that of AiVWFD and AiF10D was not calculable owing to the small number of patients. AiF13D and AiF8D were often idiopathic, whereas AiVWFD was often associated with plasma cell neoplasms. Epistaxis was a characteristic symptom of AiVWFD, intramuscular bleeding was frequent in AiF13D and AiF8D, and subcutaneous bleeding (purpura) was frequent in AiF13D and AiF10D, although none were specific to any one disease. Differential diagnosis cannot be made based on bleeding symptoms alone; therefore, rapid and accurate testing is mandatory. Definitive diagnosis of AiCFD necessitates identifying the presence of coagulation factor “inhibitors” and/or “autoantibodies.” Therefore, these tests should be performed upon unexplained severe acquired coagulation factor deficiencies. The mainstay of treatment for AiCFD was hemostatic therapy and autoantibody eradication therapy, which included the replacement of coagulation factors or “bypass” agents and administration of immunosuppressants. The rate of hemorrhagic death was high in AiF13D (13%), followed by AiF5D (7%) and Ai10D (5%); therefore, early diagnosis and optimal treatment are essential for AiCFDs. Given the unknown long-term prognosis, “intractable disease platform registries” have begun to accumulate in Japan.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1787188

    Among the acquired coagulation factor deficiencies, autoimmune coagulation factor deficiencies (AiCFD) are rare and result from autoantibody production against coagulation factors. In Japan, a nationwide survey on AiCFD has been conducted since 2009. Autoimmune factor XIII, factor VIII, von Willebrand factor, factor V, and factor X deficiencies (AiF13D, AiF8D, AiVWFD, AiF5D, and AiF10D, respectively) have been enacted as “designated intractable disease-282.” The incidence of AiF8D, AiF13D, and AiF5D was 1.83, 0.044, and 0.038 per million people/year, respectively, whereas that of AiVWFD and AiF10D was not calculable owing to the small number of patients. AiF13D and AiF8D were often idiopathic, whereas AiVWFD was often associated with plasma cell neoplasms. Epistaxis was a characteristic symptom of AiVWFD, intramuscular bleeding was frequent in AiF13D and AiF8D, and subcutaneous bleeding (purpura) was frequent in AiF13D and AiF10D, although none were specific to any one disease. Differential diagnosis cannot be made based on bleeding symptoms alone; therefore, rapid and accurate testing is mandatory. Definitive diagnosis of AiCFD necessitates identifying the presence of coagulation factor “inhibitors” and/or “autoantibodies.” Therefore, these tests should be performed upon unexplained severe acquired coagulation factor deficiencies. The mainstay of treatment for AiCFD was hemostatic therapy and autoantibody eradication therapy, which included the replacement of coagulation factors or “bypass” agents and administration of immunosuppressants. The rate of hemorrhagic death was high in AiF13D (13%), followed by AiF5D (7%) and Ai10D (5%); therefore, early diagnosis and optimal treatment are essential for AiCFDs. Given the unknown long-term prognosis, “intractable disease platform registries” have begun to accumulate in Japan.
    [...]

    Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Diagnosis and Treatment of Autoimmune Acquired Coagulation Factor Deficiencies: An Evidence-Based Review of Japanese PracticeIchinose, AkitadaOsaki, TsukasaSouri, MasayoshiDOI:10.1055/s-0044-1787188Semin Thromb Hemost ; : -2024-06-12T14:01:58+01:00Seminars in Thrombosis and Hemostasis2024-06-12T14:01:58+01:00eFirst
    Review Article
    10.1055/s-0044-1787188http://dx.doi.org/10.1055/s-0044-1787188
    Strategies for Performing Factor Assays in the Presence of Emicizumab or Other Novel/Emerging Hemostatic Agentshttp://dx.doi.org/10.1055/s-0044-1787189For several decades, therapeutic options for inherited deficiencies of factor VIII or IX (hemophilia A or B, respectively) have largely been the replacement of the missing clotting factor with plasma-derived or recombinant products. Hemostasis laboratories use standard activated partial thromboplastin time (aPTT)-based clotting or chromogenic assays to monitor plasma factor levels to guide therapy. The emergence in the past 10 years of extended half-life replacement products and other novel therapies for hemophilia has led to a reappraisal of assay suitability, with studies of product measurement showing some existing assay types or reagents to be unsuitable for some products. The hemostasis laboratory must adapt to the changing landscape by adding new assays or modifying existing assays to ensure accurate results for product measurement. These strategies include switching from a chromogenic assay to a clotting assay, or vice versa, changing an aPTT reagent brand, or introducing product specific calibrators. This article evaluates the effects of some of the newer treatment options on the laboratory testing of factor levels and related assays.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1787189

    For several decades, therapeutic options for inherited deficiencies of factor VIII or IX (hemophilia A or B, respectively) have largely been the replacement of the missing clotting factor with plasma-derived or recombinant products. Hemostasis laboratories use standard activated partial thromboplastin time (aPTT)-based clotting or chromogenic assays to monitor plasma factor levels to guide therapy. The emergence in the past 10 years of extended half-life replacement products and other novel therapies for hemophilia has led to a reappraisal of assay suitability, with studies of product measurement showing some existing assay types or reagents to be unsuitable for some products. The hemostasis laboratory must adapt to the changing landscape by adding new assays or modifying existing assays to ensure accurate results for product measurement. These strategies include switching from a chromogenic assay to a clotting assay, or vice versa, changing an aPTT reagent brand, or introducing product specific calibrators. This article evaluates the effects of some of the newer treatment options on the laboratory testing of factor levels and related assays.
    [...]

    Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Strategies for Performing Factor Assays in the Presence of Emicizumab or Other Novel/Emerging Hemostatic AgentsKershaw, GeoffreyDOI:10.1055/s-0044-1787189Semin Thromb Hemost ; : -2024-06-12T13:15:55+01:00Seminars in Thrombosis and Hemostasis2024-06-12T13:15:55+01:00eFirst
    Review Article
    10.1055/s-0044-1787189http://dx.doi.org/10.1055/s-0044-1787189
    Gene Therapy for Hemophilia B: Achievements, Open Issues, and Perspectiveshttp://dx.doi.org/10.1055/s-0044-1787190Hemophilia B is the first bleeding disorder for which gene therapy clinical programs began. Presently, adenovirus-associated vectors represent the best means to deliver the transgene, and their administration by intravenous route has been used in recent clinical trials. The natural occurring factor IX (FIX) Padua variant, which allows for a 5- to 8-fold higher activity of FIX, while maintaining a normal protein concentration, was subsequently used to enhance the level of transgene expression. All the recent trials using this variant showed good results, and accumulating data suggest that long-term expression durability could be maintained at a significant hemostatic level. However, the risk of loss of transgene expression associated to immune response with liver enzymes elevation remains a concern, especially as to the efficacy and duration of immunosuppressive treatment. Notwithstanding this limitation, the results of clinical trials suggest that gene therapy in hemophilia B has the potential to provide long-term benefits with sustained factor activity levels predicted to last several years in many patients.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1787190

    Hemophilia B is the first bleeding disorder for which gene therapy clinical programs began. Presently, adenovirus-associated vectors represent the best means to deliver the transgene, and their administration by intravenous route has been used in recent clinical trials. The natural occurring factor IX (FIX) Padua variant, which allows for a 5- to 8-fold higher activity of FIX, while maintaining a normal protein concentration, was subsequently used to enhance the level of transgene expression. All the recent trials using this variant showed good results, and accumulating data suggest that long-term expression durability could be maintained at a significant hemostatic level. However, the risk of loss of transgene expression associated to immune response with liver enzymes elevation remains a concern, especially as to the efficacy and duration of immunosuppressive treatment. Notwithstanding this limitation, the results of clinical trials suggest that gene therapy in hemophilia B has the potential to provide long-term benefits with sustained factor activity levels predicted to last several years in many patients.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Gene Therapy for Hemophilia B: Achievements, Open Issues, and PerspectivesCastaman, GiancarloMiesbach, WolfgangDOI:10.1055/s-0044-1787190Semin Thromb Hemost ; : -2024-05-31T14:07:11+01:00Seminars in Thrombosis and Hemostasis2024-05-31T14:07:11+01:00eFirst
    Review Article
    10.1055/s-0044-1787190http://dx.doi.org/10.1055/s-0044-1787190
    Pleiotropic Effects of Heparin and its Monitoring in the Clinical Practicehttp://dx.doi.org/10.1055/s-0044-1786990Unfractionated heparin (UFH) was uncovered in 1916, has been used as an anticoagulant since 1935, and has been listed in the World Health Organization's Model List of Essential Medicines. Despite the availability of many other anticoagulants, the use of heparin (either low molecular weight heparin [LMWH] or UFH) is still substantial. Heparin has pleotropic effects including anticoagulant and several nonanticoagulant properties such as antiproliferative, anti-inflammatory activity, and anticomplement effects. Although UFH has been widely replaced by LMWH, UFH is still the preferred anticoagulant of choice for patients undergoing cardiopulmonary bypass surgery, extracorporeal membrane oxygenation, and patients with high-risk mechanical cardiac valves requiring temporary bridging with a parenteral anticoagulant. UFH is a highly negatively charged molecule and binds many positively charged molecules, hence has unpredictable pharmacokinetics, and variable anticoagulant effect on an individual patient basis. Therefore, anticoagulant effects of UFH may not be proportional to the dose of UFH given to any individual patient. In this review, we discuss the anticoagulant and nonanticoagulant activities of UFH, differences between UFH and LMWH, when to use UFH, different methods of monitoring the anticoagulant effects of UFH (including activated partial thromboplastin time, heparin anti-Xa activity level, and activated clotting time), while discussing pros and cons related to each method and comparison of clinical outcomes in patients treated with UFH monitored with different methods based on available evidence.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1786990

    Unfractionated heparin (UFH) was uncovered in 1916, has been used as an anticoagulant since 1935, and has been listed in the World Health Organization's Model List of Essential Medicines. Despite the availability of many other anticoagulants, the use of heparin (either low molecular weight heparin [LMWH] or UFH) is still substantial. Heparin has pleotropic effects including anticoagulant and several nonanticoagulant properties such as antiproliferative, anti-inflammatory activity, and anticomplement effects. Although UFH has been widely replaced by LMWH, UFH is still the preferred anticoagulant of choice for patients undergoing cardiopulmonary bypass surgery, extracorporeal membrane oxygenation, and patients with high-risk mechanical cardiac valves requiring temporary bridging with a parenteral anticoagulant. UFH is a highly negatively charged molecule and binds many positively charged molecules, hence has unpredictable pharmacokinetics, and variable anticoagulant effect on an individual patient basis. Therefore, anticoagulant effects of UFH may not be proportional to the dose of UFH given to any individual patient. In this review, we discuss the anticoagulant and nonanticoagulant activities of UFH, differences between UFH and LMWH, when to use UFH, different methods of monitoring the anticoagulant effects of UFH (including activated partial thromboplastin time, heparin anti-Xa activity level, and activated clotting time), while discussing pros and cons related to each method and comparison of clinical outcomes in patients treated with UFH monitored with different methods based on available evidence.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Pleiotropic Effects of Heparin and its Monitoring in the Clinical PracticeArachchillage, Deepa J.Kitchen, SteveDOI:10.1055/s-0044-1786990Semin Thromb Hemost ; : -2024-05-29T09:56:16+01:00Seminars in Thrombosis and Hemostasis2024-05-29T09:56:16+01:00eFirst
    Review Article
    10.1055/s-0044-1786990http://dx.doi.org/10.1055/s-0044-1786990
    Contact Activation: Where Thrombosis and Hemostasis Meet on a Foreign Surface, Plus a Mini-editorial Compilation (“Part XVI”)http://dx.doi.org/10.1055/s-0044-1786751Semin Thromb Hemost
    DOI: 10.1055/s-0044-1786751



    Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

    Article in Thieme eJournals:
    Table of contents  |  Full text

    ]]>
    Contact Activation: Where Thrombosis and Hemostasis Meet on a Foreign Surface, Plus a Mini-editorial Compilation (“Part XVI”)Vu, Helen H.McCarty, Owen J.T.Favaloro, Emmanuel J.DOI:10.1055/s-0044-1786751Semin Thromb Hemost ; : -2024-05-17T11:26:50+01:00Seminars in Thrombosis and Hemostasis2024-05-17T11:26:50+01:00eFirst
    Preface
    10.1055/s-0044-1786751http://dx.doi.org/10.1055/s-0044-1786751
    Heparins May Not Be the Optimal Anticoagulants for Sepsis and Sepsis-Associated Disseminated Intravascular Coagulationhttp://dx.doi.org/10.1055/s-0044-1786754Historically, heparin has had the longest historical use as an anticoagulant and continues this day to be the primary therapeutic option for preventing thrombosis and thromboembolism in critically ill hospitalized patients. Heparin is also used to treat sepsis and sepsis-associated disseminated intravascular coagulation (DIC) in various countries. However, the efficacy and safety of heparin for this indication remains controversial, as adequately powered randomized clinical studies have not demonstrated as yet a survival benefit in sepsis and sepsis-associated DIC, despite meta-analyses and propensity analyses reporting improved outcomes without increasing bleeding risk. Further, activated protein C and recombinant thrombomodulin showed greater improvements in outcomes compared with heparin, although these effects were inconclusive. In summary, further research is warranted, despite the ongoing clinical use of heparin for sepsis and sepsis-associated DIC. Based on Japanese guidelines, antithrombin or recombinant thrombomodulin may be a preferable choice if they are accessible.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1786754

    Historically, heparin has had the longest historical use as an anticoagulant and continues this day to be the primary therapeutic option for preventing thrombosis and thromboembolism in critically ill hospitalized patients. Heparin is also used to treat sepsis and sepsis-associated disseminated intravascular coagulation (DIC) in various countries. However, the efficacy and safety of heparin for this indication remains controversial, as adequately powered randomized clinical studies have not demonstrated as yet a survival benefit in sepsis and sepsis-associated DIC, despite meta-analyses and propensity analyses reporting improved outcomes without increasing bleeding risk. Further, activated protein C and recombinant thrombomodulin showed greater improvements in outcomes compared with heparin, although these effects were inconclusive. In summary, further research is warranted, despite the ongoing clinical use of heparin for sepsis and sepsis-associated DIC. Based on Japanese guidelines, antithrombin or recombinant thrombomodulin may be a preferable choice if they are accessible.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Heparins May Not Be the Optimal Anticoagulants for Sepsis and Sepsis-Associated Disseminated Intravascular CoagulationIba, ToshiakiHelms, JulieTotoki, TakaakiLevy, Jerrold H.DOI:10.1055/s-0044-1786754Semin Thromb Hemost ; : -2024-05-11T10:09:33+01:00Seminars in Thrombosis and Hemostasis2024-05-11T10:09:33+01:00eFirst
    Commentary
    10.1055/s-0044-1786754http://dx.doi.org/10.1055/s-0044-1786754
    The Next, Next-Generation of Sequencing, Promising to Boost Research and Clinical Practicehttp://dx.doi.org/10.1055/s-0044-1786756Semin Thromb Hemost
    DOI: 10.1055/s-0044-1786756



    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Full text

    ]]>
    The Next, Next-Generation of Sequencing, Promising to Boost Research and Clinical PracticeKumar, Kishore R.Cowley, Mark J.Davis, Ryan L.DOI:10.1055/s-0044-1786756Semin Thromb Hemost ; : -2024-05-11T10:09:30+01:00Seminars in Thrombosis and Hemostasis2024-05-11T10:09:30+01:00eFirst
    Commentary
    10.1055/s-0044-1786756http://dx.doi.org/10.1055/s-0044-1786756
    Prone Position and the Risk of Venous Thrombosis in COVID-19 Patients with Respiratory Failurehttp://dx.doi.org/10.1055/s-0044-1786735Semin Thromb Hemost
    DOI: 10.1055/s-0044-1786735



    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Full text

    ]]>
    Prone Position and the Risk of Venous Thrombosis in COVID-19 Patients with Respiratory FailureLippi, GiuseppeMattiuzzi, CamillaFavaloro, Emmanuel J.DOI:10.1055/s-0044-1786735Semin Thromb Hemost ; : -2024-05-11T10:09:31+01:00Seminars in Thrombosis and Hemostasis2024-05-11T10:09:31+01:00eFirst
    Commentary
    10.1055/s-0044-1786735http://dx.doi.org/10.1055/s-0044-1786735
    Evaluating Diagnostic Algorithms for Heparin-Induced Thrombocytopenia using Two Combined Automated Rapid Immunoassayshttp://dx.doi.org/10.1055/s-0044-1786749Heparin-induced thrombocytopenia (HIT) is an autoimmune disorder caused by antibodies against platelet factor 4 (PF4) and heparin complexes. Rapid immunoassays (IAs) for detection of these antibodies mark a milestone in HIT diagnosis, despite a higher false-positive rate compared with functional platelet-activation assays. However, combining different rapid IAs may help to improve their diagnostic specificity. Here, we compared the individual performance of the latex immunoturbidimetric assay (LIA; HemosIL HIT-Ab [PF4-H]; sensitivity 91.7%, specificity 68.4%) and chemiluminescence immunoassay (CLIA; HemosIL AcuStarHIT-Ab [PF4-H]; sensitivity 92.4%, specificity 85.8%) with their combined performance using two unique diagnostic algorithms in a single prospective cohort of suspected HIT patients. Using the simultaneous algorithm adapted from Warkentin et al, the combined LIA–CLIA had a sensitivity of 99.0% and specificity of 64.3%. The sequential algorithm adapted from Rittener-Ruff et al was applied in two theoretical scenarios to reflect real-world circumstances in diagnostic laboratories where access to clinical information is limited: (1) assuming all patients had an intermediate 4Ts score and (2) assuming all patients had a high 4Ts score. This algorithm correctly predicted HIT in 94.5% (high 4Ts) and 96.0% (intermediate 4Ts) and excluded HIT in 82.6% (high 4Ts) and 80.1% (intermediate 4Ts) of patients in either scenario, respectively. Although both combined algorithms improved diagnostic performance of individual IAs, the simultaneous algorithm showed fewer false predictions (7.9%) than the sequential algorithm (intermediate 4Ts: 37.6% and high 4Ts: 41.5%) and proved more practical as it does not rely on physician evaluations. Our findings highlight the importance of accounting for clinician and interlaboratory variability when evaluating diagnostic tests for HIT.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1786749

    Heparin-induced thrombocytopenia (HIT) is an autoimmune disorder caused by antibodies against platelet factor 4 (PF4) and heparin complexes. Rapid immunoassays (IAs) for detection of these antibodies mark a milestone in HIT diagnosis, despite a higher false-positive rate compared with functional platelet-activation assays. However, combining different rapid IAs may help to improve their diagnostic specificity. Here, we compared the individual performance of the latex immunoturbidimetric assay (LIA; HemosIL HIT-Ab [PF4-H]; sensitivity 91.7%, specificity 68.4%) and chemiluminescence immunoassay (CLIA; HemosIL AcuStarHIT-Ab [PF4-H]; sensitivity 92.4%, specificity 85.8%) with their combined performance using two unique diagnostic algorithms in a single prospective cohort of suspected HIT patients. Using the simultaneous algorithm adapted from Warkentin et al, the combined LIA–CLIA had a sensitivity of 99.0% and specificity of 64.3%. The sequential algorithm adapted from Rittener-Ruff et al was applied in two theoretical scenarios to reflect real-world circumstances in diagnostic laboratories where access to clinical information is limited: (1) assuming all patients had an intermediate 4Ts score and (2) assuming all patients had a high 4Ts score. This algorithm correctly predicted HIT in 94.5% (high 4Ts) and 96.0% (intermediate 4Ts) and excluded HIT in 82.6% (high 4Ts) and 80.1% (intermediate 4Ts) of patients in either scenario, respectively. Although both combined algorithms improved diagnostic performance of individual IAs, the simultaneous algorithm showed fewer false predictions (7.9%) than the sequential algorithm (intermediate 4Ts: 37.6% and high 4Ts: 41.5%) and proved more practical as it does not rely on physician evaluations. Our findings highlight the importance of accounting for clinician and interlaboratory variability when evaluating diagnostic tests for HIT.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Evaluating Diagnostic Algorithms for Heparin-Induced Thrombocytopenia using Two Combined Automated Rapid ImmunoassaysBissola, Anna-LiseZhang, YiCranstone, MadisonMoore, Jane C.Warkentin, Theodore E.Arnold, Donald M.Nazy, IshacDOI:10.1055/s-0044-1786749Semin Thromb Hemost ; : -2024-05-11T10:09:32+01:00Seminars in Thrombosis and Hemostasis2024-05-11T10:09:32+01:00eFirst
    Review Article
    10.1055/s-0044-1786749http://dx.doi.org/10.1055/s-0044-1786749
    Nonsevere Hemophilia: The Need for a Renewed Focus and Improved Outcomeshttp://dx.doi.org/10.1055/s-0044-1786358People with nonsevere hemophilia (PWNSH) are phenotypically more diverse than those with severe hemophilia. Perceptions relating to a “nonsevere” phenotype have contributed to fewer research initiatives, fewer guidelines on optimal management, and a lack of standards for surveillance and clinical assessment for affected individuals. In many cases, episodes of abnormal bleeding could, if investigated, have led to earlier diagnosis. Furthermore, the major recent developments in therapy for hemophilia have largely focused on severe disease and, as a group, PWNSH have not been included in many key clinical trials. Benefiting people with severe disease, innovative replacement therapies have generally targeted factor levels that are above those present in a large proportion of PWNSH. Therapeutic advances can lead to improvement in phenotype for people with severe hemophilia over that currently experienced by many PWNSH. As a result, we are approaching a point where PWNSH may, in many countries, have a higher risk of bleeding and restriction in lifestyle than those with severe disease but with more limited therapeutic options. Given the multiple major advances in treatment for people with hemophilia, it is timely to review the aspects of nonsevere disease, to ensure equity in care and management for all individuals with this condition.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1786358

    People with nonsevere hemophilia (PWNSH) are phenotypically more diverse than those with severe hemophilia. Perceptions relating to a “nonsevere” phenotype have contributed to fewer research initiatives, fewer guidelines on optimal management, and a lack of standards for surveillance and clinical assessment for affected individuals. In many cases, episodes of abnormal bleeding could, if investigated, have led to earlier diagnosis. Furthermore, the major recent developments in therapy for hemophilia have largely focused on severe disease and, as a group, PWNSH have not been included in many key clinical trials. Benefiting people with severe disease, innovative replacement therapies have generally targeted factor levels that are above those present in a large proportion of PWNSH. Therapeutic advances can lead to improvement in phenotype for people with severe hemophilia over that currently experienced by many PWNSH. As a result, we are approaching a point where PWNSH may, in many countries, have a higher risk of bleeding and restriction in lifestyle than those with severe disease but with more limited therapeutic options. Given the multiple major advances in treatment for people with hemophilia, it is timely to review the aspects of nonsevere disease, to ensure equity in care and management for all individuals with this condition.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  open access Full text

    ]]>
    Nonsevere Hemophilia: The Need for a Renewed Focus and Improved OutcomesDolan, GerardFijnvandraat, KarinLenting, Peter J.Catarino, CristinaLavin, MichelleDOI:10.1055/s-0044-1786358Semin Thromb Hemost ; : -2024-05-11T10:09:34+01:00Seminars in Thrombosis and Hemostasis2024-05-11T10:09:34+01:00eFirst
    Review Article
    10.1055/s-0044-1786358http://dx.doi.org/10.1055/s-0044-1786358
    Thrombophilia Screening: Not So Straightforwardhttp://dx.doi.org/10.1055/s-0044-1786807Although inherited thrombophilias are lifelong risk factors for a first thrombotic episode, progression to thrombosis is multifactorial and not all individuals with inherited thrombophilia develop thrombosis in their lifetimes. Consequently, indiscriminate screening in patients with idiopathic thrombosis is not recommended, since presence of a thrombophilia does not necessarily predict recurrence or influence management, and testing should be selective. It follows that a decision to undertake laboratory detection of thrombophilia should be aligned with a concerted effort to identify any significant abnormalities, because it will inform patient management. Deficiencies of antithrombin and protein C are rare and usually determined using phenotypic assays assessing biological activities, whereas protein S deficiency (also rare) is commonly detected with antigenic assays for the free form of protein S since available activity assays are considered to lack specificity. In each case, no single phenotypic assay is capable of detecting every deficiency, because the various mutations express different molecular characteristics, rendering thrombophilia screening repertoires employing one assay per potential deficiency, of limited effectiveness. Activated protein C resistance (APCR) is more common than discrete deficiencies of antithrombin, protein C, and protein S and also often detected initially with phenotypic assays; however, some centres perform only genetic analysis for factor V Leiden, as this is responsible for most cases of hereditary APCR, accepting that acquired APCR and rare F5 mutations conferring APCR will go undetected if only factor V Leiden is evaluated. All phenotypic assays have interferences and limitations, which must be factored into decisions about if, and when, to test, and be given consideration in the laboratory during assay performance and interpretation. This review looks in detail at performance and limitations of routine phenotypic thrombophilia assays.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1786807

    Although inherited thrombophilias are lifelong risk factors for a first thrombotic episode, progression to thrombosis is multifactorial and not all individuals with inherited thrombophilia develop thrombosis in their lifetimes. Consequently, indiscriminate screening in patients with idiopathic thrombosis is not recommended, since presence of a thrombophilia does not necessarily predict recurrence or influence management, and testing should be selective. It follows that a decision to undertake laboratory detection of thrombophilia should be aligned with a concerted effort to identify any significant abnormalities, because it will inform patient management. Deficiencies of antithrombin and protein C are rare and usually determined using phenotypic assays assessing biological activities, whereas protein S deficiency (also rare) is commonly detected with antigenic assays for the free form of protein S since available activity assays are considered to lack specificity. In each case, no single phenotypic assay is capable of detecting every deficiency, because the various mutations express different molecular characteristics, rendering thrombophilia screening repertoires employing one assay per potential deficiency, of limited effectiveness. Activated protein C resistance (APCR) is more common than discrete deficiencies of antithrombin, protein C, and protein S and also often detected initially with phenotypic assays; however, some centres perform only genetic analysis for factor V Leiden, as this is responsible for most cases of hereditary APCR, accepting that acquired APCR and rare F5 mutations conferring APCR will go undetected if only factor V Leiden is evaluated. All phenotypic assays have interferences and limitations, which must be factored into decisions about if, and when, to test, and be given consideration in the laboratory during assay performance and interpretation. This review looks in detail at performance and limitations of routine phenotypic thrombophilia assays.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Thrombophilia Screening: Not So StraightforwardMoore, Gary W.DOI:10.1055/s-0044-1786807Semin Thromb Hemost ; : -2024-05-11T10:09:32+01:00Seminars in Thrombosis and Hemostasis2024-05-11T10:09:32+01:00eFirst
    Review Article
    10.1055/s-0044-1786807http://dx.doi.org/10.1055/s-0044-1786807
    Next-Generation Sequencing and Emerging Technologies*http://dx.doi.org/10.1055/s-0044-1786397Genetic sequencing technologies are evolving at a rapid pace with major implications for research and clinical practice. In this review, the authors provide an updated overview of next-generation sequencing (NGS) and emerging methodologies. NGS has tremendously improved sequencing output while being more time and cost-efficient in comparison to Sanger sequencing. The authors describe short-read sequencing approaches, such as sequencing by synthesis, ion semiconductor sequencing, and nanoball sequencing. Third-generation long-read sequencing now promises to overcome many of the limitations of short-read sequencing, such as the ability to reliably resolve repeat sequences and large genomic rearrangements. By combining complementary methods with massively parallel DNA sequencing, a greater insight into the biological context of disease mechanisms is now possible. Emerging methodologies, such as advances in nanopore technology, in situ nucleic acid sequencing, and microscopy-based sequencing, will continue the rapid evolution of this area. These new technologies hold many potential applications for hematological disorders, with the promise of precision and personalized medical care in the future.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1786397

    Genetic sequencing technologies are evolving at a rapid pace with major implications for research and clinical practice. In this review, the authors provide an updated overview of next-generation sequencing (NGS) and emerging methodologies. NGS has tremendously improved sequencing output while being more time and cost-efficient in comparison to Sanger sequencing. The authors describe short-read sequencing approaches, such as sequencing by synthesis, ion semiconductor sequencing, and nanoball sequencing. Third-generation long-read sequencing now promises to overcome many of the limitations of short-read sequencing, such as the ability to reliably resolve repeat sequences and large genomic rearrangements. By combining complementary methods with massively parallel DNA sequencing, a greater insight into the biological context of disease mechanisms is now possible. Emerging methodologies, such as advances in nanopore technology, in situ nucleic acid sequencing, and microscopy-based sequencing, will continue the rapid evolution of this area. These new technologies hold many potential applications for hematological disorders, with the promise of precision and personalized medical care in the future.
    [...]

    Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Next-Generation Sequencing and Emerging Technologies*Kumar, Kishore R.Cowley, Mark J.Davis, Ryan L.DOI:10.1055/s-0044-1786397Semin Thromb Hemost ; : -2024-05-01T07:51:06+01:00Seminars in Thrombosis and Hemostasis2024-05-01T07:51:06+01:00eFirst
    Historical Paper
    10.1055/s-0044-1786397http://dx.doi.org/10.1055/s-0044-1786397
    Platelet Physiology http://dx.doi.org/10.1055/s-0044-1786387Platelets are the smallest blood cells, numbering 150 to 350 × 109/L in healthy individuals. The ability of activated platelets to adhere to an injured vessel wall and form aggregates was first described in the 19th century. Besides their long-established roles in thrombosis and hemostasis, platelets are increasingly recognized as pivotal players in numerous other pathophysiological processes including inflammation and atherogenesis, antimicrobial host defense, and tumor growth and metastasis. Consequently, profound knowledge of platelet structure and function is becoming more important in research and in many fields of modern medicine. This review provides an overview of platelet physiology focusing particularly on the structure, granules, surface glycoproteins, and activation pathways of platelets.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1786387

    Platelets are the smallest blood cells, numbering 150 to 350 × 109/L in healthy individuals. The ability of activated platelets to adhere to an injured vessel wall and form aggregates was first described in the 19th century. Besides their long-established roles in thrombosis and hemostasis, platelets are increasingly recognized as pivotal players in numerous other pathophysiological processes including inflammation and atherogenesis, antimicrobial host defense, and tumor growth and metastasis. Consequently, profound knowledge of platelet structure and function is becoming more important in research and in many fields of modern medicine. This review provides an overview of platelet physiology focusing particularly on the structure, granules, surface glycoproteins, and activation pathways of platelets.
    [...]

    Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Platelet Physiology Gremmel, ThomasFrelinger, Andrew L.Michelson, Alan D.DOI:10.1055/s-0044-1786387Semin Thromb Hemost ; : -2024-04-23T10:58:27+01:00Seminars in Thrombosis and Hemostasis2024-04-23T10:58:27+01:00eFirst
    Historical Paper
    10.1055/s-0044-1786387http://dx.doi.org/10.1055/s-0044-1786387
    Heterozygous ITGA2B Phe1024 Deletion Associated with Abnormal αIIbβ3 Function in a Patient with Congenital Thrombocytopeniahttp://dx.doi.org/10.1055/s-0044-1785655Semin Thromb Hemost
    DOI: 10.1055/s-0044-1785655



    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Full text

    ]]>
    Heterozygous ITGA2B Phe1024 Deletion Associated with Abnormal αIIbβ3 Function in a Patient with Congenital ThrombocytopeniaWang, BinTang, NingHou, HongyanChen, JunkunWang, XiongLi, JiaoyuanDOI:10.1055/s-0044-1785655Semin Thromb Hemost ; : -2024-04-11T13:10:44+01:00Seminars in Thrombosis and Hemostasis2024-04-11T13:10:44+01:00eFirst
    Letter to the Editor
    10.1055/s-0044-1785655http://dx.doi.org/10.1055/s-0044-1785655
    Heparin-induced Thrombocytopenia with Thrombosis in COVID-19 versus Vaccine-induced Immune Thrombocytopenia and Thrombosis in the United Kingdomhttp://dx.doi.org/10.1055/s-0044-1785484Semin Thromb Hemost
    DOI: 10.1055/s-0044-1785484



    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Full text

    ]]>
    Heparin-induced Thrombocytopenia with Thrombosis in COVID-19 versus Vaccine-induced Immune Thrombocytopenia and Thrombosis in the United KingdomArachchillage, Deepa J.Rajakaruna, IndikaMakris, MikeLaffan, MikeDOI:10.1055/s-0044-1785484Semin Thromb Hemost ; : -2024-04-09T06:48:07+01:00Seminars in Thrombosis and Hemostasis2024-04-09T06:48:07+01:00eFirst
    Letter to the Editor
    10.1055/s-0044-1785484http://dx.doi.org/10.1055/s-0044-1785484
    Gene Therapy in Hemophilia A: Achievements, Challenges, and Perspectiveshttp://dx.doi.org/10.1055/s-0044-1785483Strides in advancements of care of persons with hemophilia include development of long-acting factor replacement therapies, novel substitution and hemostatic rebalancing agents, and most recently approved gene therapy. Several decades of preclinical and clinical trials have led to development of adeno-associated viral (AAV) vector-mediated gene transfer for endogenous production of factor VIII (FVIII) in hemophilia A (HA). Only one gene therapy product for HA (valoctocogene roxaparvovec) has been approved by regulatory authorities. Results of valoctocogene roxaparvovec trial show significant improvement in bleeding rates and use of factor replacement therapy; however, sustainability and duration of response show variability with overall decline in FVIII expression over time. Further challenges include untoward adverse effects involving liver toxicity requiring immunosuppression and development of neutralizing antibodies to AAV vector rendering future doses ineffective. Real-life applicability of gene therapy for HA will require appropriate patient screening, infrastructure setup, long-term monitoring including data collection of patient-reported outcomes and innovative payment schemes. This review article highlights the success and development of HA gene therapy trials, challenges including adverse outcomes and variability of response, and perspectives on approach to gene therapy including shared decision-making and need for future strategies to overcome the several unmet needs.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1785483

    Strides in advancements of care of persons with hemophilia include development of long-acting factor replacement therapies, novel substitution and hemostatic rebalancing agents, and most recently approved gene therapy. Several decades of preclinical and clinical trials have led to development of adeno-associated viral (AAV) vector-mediated gene transfer for endogenous production of factor VIII (FVIII) in hemophilia A (HA). Only one gene therapy product for HA (valoctocogene roxaparvovec) has been approved by regulatory authorities. Results of valoctocogene roxaparvovec trial show significant improvement in bleeding rates and use of factor replacement therapy; however, sustainability and duration of response show variability with overall decline in FVIII expression over time. Further challenges include untoward adverse effects involving liver toxicity requiring immunosuppression and development of neutralizing antibodies to AAV vector rendering future doses ineffective. Real-life applicability of gene therapy for HA will require appropriate patient screening, infrastructure setup, long-term monitoring including data collection of patient-reported outcomes and innovative payment schemes. This review article highlights the success and development of HA gene therapy trials, challenges including adverse outcomes and variability of response, and perspectives on approach to gene therapy including shared decision-making and need for future strategies to overcome the several unmet needs.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Gene Therapy in Hemophilia A: Achievements, Challenges, and PerspectivesBala, Natasha S.Thornburg, Courtney D.DOI:10.1055/s-0044-1785483Semin Thromb Hemost ; : -2024-04-08T06:36:42+01:00Seminars in Thrombosis and Hemostasis2024-04-08T06:36:42+01:00eFirst
    Review Article
    10.1055/s-0044-1785483http://dx.doi.org/10.1055/s-0044-1785483
    The Role of Myeloid Cells in Thromboinflammatory Diseasehttp://dx.doi.org/10.1055/s-0044-1782660Inflammation contributes to the development of thrombosis, but the mechanistic basis for this association remains poorly understood. Innate immune responses and coagulation pathways are activated in parallel following infection or injury, and represent an important host defense mechanism to limit pathogen spread in the bloodstream. However, dysregulated proinflammatory activity is implicated in the progression of venous thromboembolism and arterial thrombosis. In this review, we focus on the role of myeloid cells in propagating thromboinflammation in acute inflammatory conditions, such as sepsis and coronavirus disease 2019 (COVID-19), and chronic inflammatory conditions, such as obesity, atherosclerosis, and inflammatory bowel disease. Myeloid cells are considered key drivers of thromboinflammation via upregulated tissue factor activity, formation of neutrophil extracellular traps (NETs), contact pathway activation, and aberrant coagulation factor–mediated protease-activated receptor (PAR) signaling. We discuss how strategies to target the intersection between myeloid cell–mediated inflammation and activation of blood coagulation represent an exciting new approach to combat immunothrombosis. Specifically, repurposed anti-inflammatory drugs, immunometabolic regulators, and NETosis inhibitors present opportunities that have the potential to dampen immunothrombotic activity without interfering with hemostasis. Such therapies could have far-reaching benefits for patient care across many thromboinflammatory conditions.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1782660

    Inflammation contributes to the development of thrombosis, but the mechanistic basis for this association remains poorly understood. Innate immune responses and coagulation pathways are activated in parallel following infection or injury, and represent an important host defense mechanism to limit pathogen spread in the bloodstream. However, dysregulated proinflammatory activity is implicated in the progression of venous thromboembolism and arterial thrombosis. In this review, we focus on the role of myeloid cells in propagating thromboinflammation in acute inflammatory conditions, such as sepsis and coronavirus disease 2019 (COVID-19), and chronic inflammatory conditions, such as obesity, atherosclerosis, and inflammatory bowel disease. Myeloid cells are considered key drivers of thromboinflammation via upregulated tissue factor activity, formation of neutrophil extracellular traps (NETs), contact pathway activation, and aberrant coagulation factor–mediated protease-activated receptor (PAR) signaling. We discuss how strategies to target the intersection between myeloid cell–mediated inflammation and activation of blood coagulation represent an exciting new approach to combat immunothrombosis. Specifically, repurposed anti-inflammatory drugs, immunometabolic regulators, and NETosis inhibitors present opportunities that have the potential to dampen immunothrombotic activity without interfering with hemostasis. Such therapies could have far-reaching benefits for patient care across many thromboinflammatory conditions.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    The Role of Myeloid Cells in Thromboinflammatory DiseaseNoone, DavidPreston, Roger J.S.Rehill, Aisling M.DOI:10.1055/s-0044-1782660Semin Thromb Hemost ; : -2024-03-28T07:54:28+0100Seminars in Thrombosis and Hemostasis2024-03-28T07:54:28+0100eFirst
    Review Article
    10.1055/s-0044-1782660http://dx.doi.org/10.1055/s-0044-1782660
    Glanzmann Thrombasthenia 10 Years Later: Progress Made and Future Directionshttp://dx.doi.org/10.1055/s-0044-1782519Glanzmann thrombasthenia (GT) is the most common inherited platelet disorder (IPD) with mucocutaneous bleeding and a failure of platelets to aggregate when stimulated. The molecular cause is insufficient or defective αIIbβ3, an integrin encoded by the ITGA2B and ITGB3 genes. On activation αIIbβ3 undergoes conformational changes and binds fibrinogen (Fg) and other proteins to join platelets in the aggregate. The application of next-generation sequencing (NGS) to patients with IPDs has accelerated genotyping for GT; progress accompanied by improved mutation curation. The evaluation by NGS of variants in other hemostasis and vascular genes is a major step toward understanding why bleeding varies so much between patients. The recently discovered role for glycoprotein VI in thrombus formation, through its binding to fibrin and surface-bound Fg, may offer a mechanosensitive back-up for αIIbβ3, especially at sites of inflammation. The setting up of national networks for IPDs and GT is improving patient care. Hematopoietic stem cell therapy provides a long-term cure for severe cases; however, prophylaxis by monoclonal antibodies designed to accelerate fibrin formation at injured sites in the vasculature is a promising development. Gene therapy using lentil-virus vectors remains a future option with CRISPR/Cas9 technologies offering a promising alternative route.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1782519

    Glanzmann thrombasthenia (GT) is the most common inherited platelet disorder (IPD) with mucocutaneous bleeding and a failure of platelets to aggregate when stimulated. The molecular cause is insufficient or defective αIIbβ3, an integrin encoded by the ITGA2B and ITGB3 genes. On activation αIIbβ3 undergoes conformational changes and binds fibrinogen (Fg) and other proteins to join platelets in the aggregate. The application of next-generation sequencing (NGS) to patients with IPDs has accelerated genotyping for GT; progress accompanied by improved mutation curation. The evaluation by NGS of variants in other hemostasis and vascular genes is a major step toward understanding why bleeding varies so much between patients. The recently discovered role for glycoprotein VI in thrombus formation, through its binding to fibrin and surface-bound Fg, may offer a mechanosensitive back-up for αIIbβ3, especially at sites of inflammation. The setting up of national networks for IPDs and GT is improving patient care. Hematopoietic stem cell therapy provides a long-term cure for severe cases; however, prophylaxis by monoclonal antibodies designed to accelerate fibrin formation at injured sites in the vasculature is a promising development. Gene therapy using lentil-virus vectors remains a future option with CRISPR/Cas9 technologies offering a promising alternative route.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Glanzmann Thrombasthenia 10 Years Later: Progress Made and Future DirectionsNurden, Alan T.Nurden, PaquitaDOI:10.1055/s-0044-1782519Semin Thromb Hemost ; : -2024-03-18T06:05:25+0100Seminars in Thrombosis and Hemostasis2024-03-18T06:05:25+0100eFirst
    Review Article
    10.1055/s-0044-1782519http://dx.doi.org/10.1055/s-0044-1782519
    2024 Eberhard F. Mammen Award Announcements: Part I—Most Popular Articleshttp://dx.doi.org/10.1055/s-0044-1782197Semin Thromb Hemost
    DOI: 10.1055/s-0044-1782197



    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Full text

    ]]>
    2024 Eberhard F. Mammen Award Announcements: Part I—Most Popular ArticlesFavaloro, Emmanuel J.DOI:10.1055/s-0044-1782197Semin Thromb Hemost ; : -2024-03-08T18:04:09+0100Seminars in Thrombosis and Hemostasis2024-03-08T18:04:09+0100eFirst
    Editorial
    10.1055/s-0044-1782197http://dx.doi.org/10.1055/s-0044-1782197
    Pearls and Pitfalls in the Measurement of Direct Oral Anticoagulantshttp://dx.doi.org/10.1055/s-0044-1782196Due to their widespread use, testing for direct oral anticoagulants (DOACs) has become urgent in certain clinical situations. Screening based on widely available, rapid, and simple hemostasis assays such as prothrombin time, activated partial thromboplastin time, or even diluted Russel Viper venom time may provide sufficient evidence of “over-coagulation” and could be used “in small/peripheral/spoke laboratories” as an emergency strategy, but is not thought to be reliable for driving clinical decision making. Given their good correlation with plasma concentration, urine dipsticks may be considered a valuable alternative for emergency screening, although their performance is dependent on renal function, may vary depending on the time since the last urination, and there may be problems of interfacing with the laboratory/hospital information system. Separation methods based on liquid chromatography and mass spectrometry may be clinically questionable, since they measure the concentration rather than the actual inhibitory effect of DOACs, are relatively expensive, cumbersome and time consuming, and therefore seem unsuitable for most conditions requiring urgent clinical decision making. A proposed approach therefore involves establishing a network of routine clinical laboratories, designating a reference center where DOAC tests could be available 24/7, establishing a clear diagnostic care pathway for ordering the tests from the laboratory and standard operating procedures for performing them, the use of the diluted thrombin time for dabigatran and anti-FXa assays (drug-calibrated) for rivaroxaban, apixaban, and edoxaban, as well as providing expert advice throughout the testing process, from ordering to interpretation of results.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1782196

    Due to their widespread use, testing for direct oral anticoagulants (DOACs) has become urgent in certain clinical situations. Screening based on widely available, rapid, and simple hemostasis assays such as prothrombin time, activated partial thromboplastin time, or even diluted Russel Viper venom time may provide sufficient evidence of “over-coagulation” and could be used “in small/peripheral/spoke laboratories” as an emergency strategy, but is not thought to be reliable for driving clinical decision making. Given their good correlation with plasma concentration, urine dipsticks may be considered a valuable alternative for emergency screening, although their performance is dependent on renal function, may vary depending on the time since the last urination, and there may be problems of interfacing with the laboratory/hospital information system. Separation methods based on liquid chromatography and mass spectrometry may be clinically questionable, since they measure the concentration rather than the actual inhibitory effect of DOACs, are relatively expensive, cumbersome and time consuming, and therefore seem unsuitable for most conditions requiring urgent clinical decision making. A proposed approach therefore involves establishing a network of routine clinical laboratories, designating a reference center where DOAC tests could be available 24/7, establishing a clear diagnostic care pathway for ordering the tests from the laboratory and standard operating procedures for performing them, the use of the diluted thrombin time for dabigatran and anti-FXa assays (drug-calibrated) for rivaroxaban, apixaban, and edoxaban, as well as providing expert advice throughout the testing process, from ordering to interpretation of results.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Pearls and Pitfalls in the Measurement of Direct Oral AnticoagulantsLippi, GiuseppeFavaloro, Emmanuel J.DOI:10.1055/s-0044-1782196Semin Thromb Hemost ; : -2024-03-08T18:04:10+0100Seminars in Thrombosis and Hemostasis2024-03-08T18:04:10+0100eFirst
    Review Article
    10.1055/s-0044-1782196http://dx.doi.org/10.1055/s-0044-1782196
    Variable Performance of D-dimer Testing by Hemostasis Laboratories: The Australasian/Asia-Pacific Experiencehttp://dx.doi.org/10.1055/s-0044-1781450D-dimers represent the breakdown products of fibrin. Thus, elevated plasma D-dimers will arise following a thrombotic event, such as a deep vein thrombosis or a pulmonary embolism, and therefore, a nonelevated D-dimer is used to effectively exclude such events. D-dimers are also elevated in a range of other conditions, for example, during disseminated intravascular coagulation. D-dimer levels may also be associated with prognostic value. For example, highly raised D-dimer levels can be associated with worsening clinical features in coronavirus disease 2019. Thus, D-dimer testing represents a commonly requested hemostasis test, often performed in 24/7 laboratories. Unfortunately, D-dimer testing is neither standardized nor harmonized across manufacturers or laboratories. Indeed, considering reporting units and the magnitude of units, up to 28 different combinations may be reported by laboratories. We provide updated findings for D-dimer testing in our geographic region, using recent data from the Royal College of Pathologists of Australasia Quality Assurance Programs, an international external quality assessment program, currently with over 450 participants in the D-dimer module. Data show a wide variety of assays in use and variable outcomes in reported numerical values when assessing proficiency samples. D-dimer testing mostly comprised reagents from three main manufacturing suppliers, with a small number of users of reagents from other manufacturers. Reported results showed important differences in numerical values for the same homogeneous tested samples when normalized to a single reporting unit (e.g., mg/L). Nevertheless, despite using different test reagents and reporting, most participants uniformly identified D-dimer values as below or above a “detection” cut-off for samples that were constructed to be below or above most cut-off values. As expected, mixed findings were reported for samples containing levels around expected cut-off values. We hope that our findings, reflecting on the heterogeneity of test reagents and test data, help improve diagnostic testing for D-dimer testing and facilitate harmonization and standardization, in the future.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1781450

    D-dimers represent the breakdown products of fibrin. Thus, elevated plasma D-dimers will arise following a thrombotic event, such as a deep vein thrombosis or a pulmonary embolism, and therefore, a nonelevated D-dimer is used to effectively exclude such events. D-dimers are also elevated in a range of other conditions, for example, during disseminated intravascular coagulation. D-dimer levels may also be associated with prognostic value. For example, highly raised D-dimer levels can be associated with worsening clinical features in coronavirus disease 2019. Thus, D-dimer testing represents a commonly requested hemostasis test, often performed in 24/7 laboratories. Unfortunately, D-dimer testing is neither standardized nor harmonized across manufacturers or laboratories. Indeed, considering reporting units and the magnitude of units, up to 28 different combinations may be reported by laboratories. We provide updated findings for D-dimer testing in our geographic region, using recent data from the Royal College of Pathologists of Australasia Quality Assurance Programs, an international external quality assessment program, currently with over 450 participants in the D-dimer module. Data show a wide variety of assays in use and variable outcomes in reported numerical values when assessing proficiency samples. D-dimer testing mostly comprised reagents from three main manufacturing suppliers, with a small number of users of reagents from other manufacturers. Reported results showed important differences in numerical values for the same homogeneous tested samples when normalized to a single reporting unit (e.g., mg/L). Nevertheless, despite using different test reagents and reporting, most participants uniformly identified D-dimer values as below or above a “detection” cut-off for samples that were constructed to be below or above most cut-off values. As expected, mixed findings were reported for samples containing levels around expected cut-off values. We hope that our findings, reflecting on the heterogeneity of test reagents and test data, help improve diagnostic testing for D-dimer testing and facilitate harmonization and standardization, in the future.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Variable Performance of D-dimer Testing by Hemostasis Laboratories: The Australasian/Asia-Pacific ExperienceFavaloro, Emmanuel J.Arunachalam, SandyaDean, ElysseDOI:10.1055/s-0044-1781450Semin Thromb Hemost ; : -2024-03-01T06:31:48+0100Seminars in Thrombosis and Hemostasis2024-03-01T06:31:48+0100eFirst
    Review Article
    10.1055/s-0044-1781450http://dx.doi.org/10.1055/s-0044-1781450
    Innovative Therapies for Acquired Hemophilia Ahttp://dx.doi.org/10.1055/s-0044-1779737Acquired hemophilia A (AHA) is a rare autoimmune bleeding disorder which can be life-threatening. AHA is due to autoantibodies against coagulation factor VIII. Disease onset may be idiopathic (approximately half of the cases) or triggered by autoimmune disorders, cancers, drugs, infections, or pregnancy. Besides treating the underlying disorder, specific AHA treatments include management of bleeding and inhibitor eradication. Various first-line and second-line hemostatic and immunosuppressive agents are currently available for the management of AHA. Recently, the hemostatic drug emicizumab and the immunosuppressive drug rituximab have been the object of intense research from investigators as innovative promising therapies for AHA. This narrative review will be focused on the current status of the clinical use of these two off-label therapeutic agents in AHA.Semin Thromb Hemost
    DOI: 10.1055/s-0044-1779737

    Acquired hemophilia A (AHA) is a rare autoimmune bleeding disorder which can be life-threatening. AHA is due to autoantibodies against coagulation factor VIII. Disease onset may be idiopathic (approximately half of the cases) or triggered by autoimmune disorders, cancers, drugs, infections, or pregnancy. Besides treating the underlying disorder, specific AHA treatments include management of bleeding and inhibitor eradication. Various first-line and second-line hemostatic and immunosuppressive agents are currently available for the management of AHA. Recently, the hemostatic drug emicizumab and the immunosuppressive drug rituximab have been the object of intense research from investigators as innovative promising therapies for AHA. This narrative review will be focused on the current status of the clinical use of these two off-label therapeutic agents in AHA.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Innovative Therapies for Acquired Hemophilia AFranchini, MassimoFocosi, DanieleDOI:10.1055/s-0044-1779737Semin Thromb Hemost ; : -2024-02-23T05:48:51+0100Seminars in Thrombosis and Hemostasis2024-02-23T05:48:51+0100eFirst
    Review Article
    10.1055/s-0044-1779737http://dx.doi.org/10.1055/s-0044-1779737
    Variable Performance of Lupus Anticoagulant Testing: The Australasian/Asia-Pacific Experiencehttp://dx.doi.org/10.1055/s-0043-1776406Lupus anticoagulant (LA) is one of three tests identified as laboratory criteria for definite antiphospholipid syndrome (APS). The other two tests are anticardiolipin antibody (aCL) and anti-β2-glycoprotein I (aβ2GPI) antibody. The presence of LA is assessed using clot-based tests, while the presence of aCL and aβ2GPI is assessed by immunological assays. Since no test can be considered 100% sensitive or specific for LA, current guidelines recommend using two different clot-based assays reflecting different principles, with the dilute Russell viper venom time (dRVVT) and activated partial thromboplastin time (aPTT) recommended. Initially, LA-sensitive reagents are used to screen for LA, and then, in “screen-positive” samples, LA-“insensitive” reagents are used to confirm LA. Because LA assays are based on clot detection, anything that can interfere with fibrin clot development may affect test results. In particular, in addition to LA, the tests are also sensitive to the presence of a wide range of clinical anticoagulants, reflecting preanalytical issues for testing. We provide updated findings for LA testing in our geographic region, using recent data from the Royal College of Pathologists of Australasia Quality Assurance Programs, an international external quality assessment program with approximately 120 participants. Data show a wide variety of assays in use, especially for aPTT testing, and variable outcomes in reported numerical values with these assays when assessing proficiency samples. dRVVT testing mostly comprised reagents from three main manufacturing suppliers, which also showed differences in numerical values for the same homogeneous tested samples. Nevertheless, despite the use of different test reagents and processes, >98% of participants correctly identified LA-negative samples as LA-negative and LA-positive samples as LA positive. We hope our findings, reflecting on the heterogeneity of test processes and test data, help improve diagnostic testing for LA in the future.Semin Thromb Hemost
    DOI: 10.1055/s-0043-1776406

    Lupus anticoagulant (LA) is one of three tests identified as laboratory criteria for definite antiphospholipid syndrome (APS). The other two tests are anticardiolipin antibody (aCL) and anti-β2-glycoprotein I (aβ2GPI) antibody. The presence of LA is assessed using clot-based tests, while the presence of aCL and aβ2GPI is assessed by immunological assays. Since no test can be considered 100% sensitive or specific for LA, current guidelines recommend using two different clot-based assays reflecting different principles, with the dilute Russell viper venom time (dRVVT) and activated partial thromboplastin time (aPTT) recommended. Initially, LA-sensitive reagents are used to screen for LA, and then, in “screen-positive” samples, LA-“insensitive” reagents are used to confirm LA. Because LA assays are based on clot detection, anything that can interfere with fibrin clot development may affect test results. In particular, in addition to LA, the tests are also sensitive to the presence of a wide range of clinical anticoagulants, reflecting preanalytical issues for testing. We provide updated findings for LA testing in our geographic region, using recent data from the Royal College of Pathologists of Australasia Quality Assurance Programs, an international external quality assessment program with approximately 120 participants. Data show a wide variety of assays in use, especially for aPTT testing, and variable outcomes in reported numerical values with these assays when assessing proficiency samples. dRVVT testing mostly comprised reagents from three main manufacturing suppliers, which also showed differences in numerical values for the same homogeneous tested samples. Nevertheless, despite the use of different test reagents and processes, >98% of participants correctly identified LA-negative samples as LA-negative and LA-positive samples as LA positive. We hope our findings, reflecting on the heterogeneity of test processes and test data, help improve diagnostic testing for LA in the future.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Variable Performance of Lupus Anticoagulant Testing: The Australasian/Asia-Pacific ExperienceFavaloro, Emmanuel J.Dean, ElysseArunachalam, SandyaDOI:10.1055/s-0043-1776406Semin Thromb Hemost ; : -2023-11-15T10:16:27+0100Seminars in Thrombosis and Hemostasis2023-11-15T10:16:27+0100eFirst
    Review Article
    10.1055/s-0043-1776406http://dx.doi.org/10.1055/s-0043-1776406
    International Council for Standardization in Haematology Guidance for New Lot Verification of Coagulation Reagents, Calibrators, and Controlshttp://dx.doi.org/10.1055/s-0043-1776405The clinical laboratory uses commercial products with limited shelf life or certain expiry dates requiring frequent lot changes. Prior to implementation for clinical use, laboratories should determine the performance of the new reagent lot to ensure that there is no significant shift in reagent performance or reporting of patient data. This guideline has been written on behalf of the International Council for Standardization in Haematology (ICSH) to provide the framework and provisional guidance for clinical laboratories for evaluating and verifying the performance of new lot reagents used for coagulation testing. These ICSH Working Party consensus recommendations are based on good laboratory practice, regulatory recommendations, evidence emerged from scientific publications, and expert opinion and are meant to supplement regional standards, regulations, or requirements.Semin Thromb Hemost
    DOI: 10.1055/s-0043-1776405

    The clinical laboratory uses commercial products with limited shelf life or certain expiry dates requiring frequent lot changes. Prior to implementation for clinical use, laboratories should determine the performance of the new reagent lot to ensure that there is no significant shift in reagent performance or reporting of patient data. This guideline has been written on behalf of the International Council for Standardization in Haematology (ICSH) to provide the framework and provisional guidance for clinical laboratories for evaluating and verifying the performance of new lot reagents used for coagulation testing. These ICSH Working Party consensus recommendations are based on good laboratory practice, regulatory recommendations, evidence emerged from scientific publications, and expert opinion and are meant to supplement regional standards, regulations, or requirements.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  open access Full text

    ]]>
    International Council for Standardization in Haematology Guidance for New Lot Verification of Coagulation Reagents, Calibrators, and ControlsGosselin, Robert C.Castellone, DonnaDorgalaleh, AkbarHickey, KieronLippi, GiuseppeMoffat, KarenO'Toole, RebeccaRigano, JoeDOI:10.1055/s-0043-1776405Semin Thromb Hemost ; : -2023-11-15T10:16:26+0100Seminars in Thrombosis and Hemostasis2023-11-15T10:16:26+0100eFirst
    Review Article
    10.1055/s-0043-1776405http://dx.doi.org/10.1055/s-0043-1776405
    Erratum: Laboratory Diagnosis of Activated Protein C Resistance and Factor V Leidenhttp://dx.doi.org/10.1055/s-0043-1776324Semin Thromb Hemost
    DOI: 10.1055/s-0043-1776324



    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Full text

    ]]>
    Erratum: Laboratory Diagnosis of Activated Protein C Resistance and Factor V LeidenBahraini, MehranFazeli, AliehDorgalaleh, AkbarDOI:10.1055/s-0043-1776324Semin Thromb Hemost ; : -2023-10-25T13:33:47+01:00Seminars in Thrombosis and Hemostasis2023-10-25T13:33:47+01:00eFirst
    Erratum
    10.1055/s-0043-1776324http://dx.doi.org/10.1055/s-0043-1776324
    Internal Quality Control in Hemostasis Assayshttp://dx.doi.org/10.1055/s-0043-1774381Internal quality control (IQC) for routine and specialist hemostasis testing represents a mandatory requirement for assays offered by clinical laboratories under International Organization for Standardization, Code of Federal Regulations, and Clinical and Laboratory Standards Institute standards. The underlying principle is that regular IQC audits the analytical performance of automated, semiautomated, and manual methods. This review investigates IQC practices, including benefits, limitations, frequency per time period or batch, sources of material used, primary supplier, third party or in-house, plus troubleshooting when IQC falls outside acceptance criteria. To assess IQC practice, the UK National External Quality Assessment Scheme (NEQAS) Blood Coagulation distributed a questionnaire to 1,200 participants enrolled in our scheme that collected details of the local practices for IQC testing. We received returns from 127 centers that described their local practices for the frequency of IQC, the type of IQC material employed, acceptance criteria for IQC data, and troubleshooting protocols for IQC failures. The data collected as part of an NEQAS BC questionnaire confirmed that all the participants returning answers to the questionnaire meet the standards for regular IQC testing for the hemostasis assays they perform.Semin Thromb Hemost
    DOI: 10.1055/s-0043-1774381

    Internal quality control (IQC) for routine and specialist hemostasis testing represents a mandatory requirement for assays offered by clinical laboratories under International Organization for Standardization, Code of Federal Regulations, and Clinical and Laboratory Standards Institute standards. The underlying principle is that regular IQC audits the analytical performance of automated, semiautomated, and manual methods. This review investigates IQC practices, including benefits, limitations, frequency per time period or batch, sources of material used, primary supplier, third party or in-house, plus troubleshooting when IQC falls outside acceptance criteria. To assess IQC practice, the UK National External Quality Assessment Scheme (NEQAS) Blood Coagulation distributed a questionnaire to 1,200 participants enrolled in our scheme that collected details of the local practices for IQC testing. We received returns from 127 centers that described their local practices for the frequency of IQC, the type of IQC material employed, acceptance criteria for IQC data, and troubleshooting protocols for IQC failures. The data collected as part of an NEQAS BC questionnaire confirmed that all the participants returning answers to the questionnaire meet the standards for regular IQC testing for the hemostasis assays they perform.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Internal Quality Control in Hemostasis AssaysReilly-Stitt, ChristopherJennings, IanKitchen, SteveWalker, Isobel D.DOI:10.1055/s-0043-1774381Semin Thromb Hemost ; : -2023-09-25T13:45:59+01:00Seminars in Thrombosis and Hemostasis2023-09-25T13:45:59+01:00eFirst
    Review Article
    10.1055/s-0043-1774381http://dx.doi.org/10.1055/s-0043-1774381
    Laboratory Diagnosis of Activated Protein C Resistance and Factor V Leidenhttp://dx.doi.org/10.1055/s-0043-1770773The factor V Leiden (FVL) polymorphism is known as the most common inherited risk factor for venous thrombosis. In turn, FVL is the leading cause of an activated protein C resistance (APCR) phenotype, in which the addition of exogenous activated protein C to plasma does not result in the expected anticoagulant effect. In the routine laboratory approach to the formal diagnosis of FVL, an initial positive screening plasma-based method for APCR is often performed, and only if needed, this is followed by a confirmatory DNA-based assay for FVL. Multiple methods with accepted sensitivity and specificity for determining an APCR/FVL phenotype are commonly categorized into two separate groups: (1) screening plasma-based assays, including qualitative functional clot-based assays, for APCR, and (2) confirmatory DNA-based molecular assays, entailing several tests and platforms, including polymerase chain reaction-based and non-PCR-based techniques, for FVL. This review will describe the methodological aspects of each laboratory test and prepare suggestions on the indication of APCR and FVL testing and method selection.Semin Thromb Hemost
    DOI: 10.1055/s-0043-1770773

    The factor V Leiden (FVL) polymorphism is known as the most common inherited risk factor for venous thrombosis. In turn, FVL is the leading cause of an activated protein C resistance (APCR) phenotype, in which the addition of exogenous activated protein C to plasma does not result in the expected anticoagulant effect. In the routine laboratory approach to the formal diagnosis of FVL, an initial positive screening plasma-based method for APCR is often performed, and only if needed, this is followed by a confirmatory DNA-based assay for FVL. Multiple methods with accepted sensitivity and specificity for determining an APCR/FVL phenotype are commonly categorized into two separate groups: (1) screening plasma-based assays, including qualitative functional clot-based assays, for APCR, and (2) confirmatory DNA-based molecular assays, entailing several tests and platforms, including polymerase chain reaction-based and non-PCR-based techniques, for FVL. This review will describe the methodological aspects of each laboratory test and prepare suggestions on the indication of APCR and FVL testing and method selection.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Laboratory Diagnosis of Activated Protein C Resistance and Factor V LeidenBahraini, MehranFazeli, AliehDorgalaleh, AkbarDOI:10.1055/s-0043-1770773Semin Thromb Hemost ; : -2023-07-10T06:05:02+01:00Seminars in Thrombosis and Hemostasis2023-07-10T06:05:02+01:00eFirst
    Review Article
    10.1055/s-0043-1770773http://dx.doi.org/10.1055/s-0043-1770773
    Factor XII Structure–Function Relationshipshttp://dx.doi.org/10.1055/s-0043-1769509Factor XII (FXII), the zymogen of the protease FXIIa, contributes to pathologic processes such as bradykinin-dependent angioedema and thrombosis through its capacity to convert the homologs prekallikrein and factor XI to the proteases plasma kallikrein and factor XIa. FXII activation and FXIIa activity are enhanced when the protein binds to a surface. Here, we review recent work on the structure and enzymology of FXII with an emphasis on how they relate to pathology. FXII is a homolog of pro-hepatocyte growth factor activator (pro-HGFA). We prepared a panel of FXII molecules in which individual domains were replaced with corresponding pro-HGFA domains and tested them in FXII activation and activity assays. When in fluid phase (not surface bound), FXII and prekallikrein undergo reciprocal activation. The FXII heavy chain restricts reciprocal activation, setting limits on the rate of this process. Pro-HGFA replacements for the FXII fibronectin type 2 or kringle domains markedly accelerate reciprocal activation, indicating disruption of the normal regulatory function of the heavy chain. Surface binding also enhances FXII activation and activity. This effect is lost if the FXII first epidermal growth factor (EGF1) domain is replaced with pro-HGFA EGF1. These results suggest that FXII circulates in blood in a “closed” form that is resistant to activation. Intramolecular interactions involving the fibronectin type 2 and kringle domains maintain the closed form. FXII binding to a surface through the EGF1 domain disrupts these interactions, resulting in an open conformation that facilitates FXII activation. These observations have implications for understanding FXII contributions to diseases such as hereditary angioedema and surface-triggered thrombosis, and for developing treatments for thrombo-inflammatory disorders.Semin Thromb Hemost
    DOI: 10.1055/s-0043-1769509

    Factor XII (FXII), the zymogen of the protease FXIIa, contributes to pathologic processes such as bradykinin-dependent angioedema and thrombosis through its capacity to convert the homologs prekallikrein and factor XI to the proteases plasma kallikrein and factor XIa. FXII activation and FXIIa activity are enhanced when the protein binds to a surface. Here, we review recent work on the structure and enzymology of FXII with an emphasis on how they relate to pathology. FXII is a homolog of pro-hepatocyte growth factor activator (pro-HGFA). We prepared a panel of FXII molecules in which individual domains were replaced with corresponding pro-HGFA domains and tested them in FXII activation and activity assays. When in fluid phase (not surface bound), FXII and prekallikrein undergo reciprocal activation. The FXII heavy chain restricts reciprocal activation, setting limits on the rate of this process. Pro-HGFA replacements for the FXII fibronectin type 2 or kringle domains markedly accelerate reciprocal activation, indicating disruption of the normal regulatory function of the heavy chain. Surface binding also enhances FXII activation and activity. This effect is lost if the FXII first epidermal growth factor (EGF1) domain is replaced with pro-HGFA EGF1. These results suggest that FXII circulates in blood in a “closed” form that is resistant to activation. Intramolecular interactions involving the fibronectin type 2 and kringle domains maintain the closed form. FXII binding to a surface through the EGF1 domain disrupts these interactions, resulting in an open conformation that facilitates FXII activation. These observations have implications for understanding FXII contributions to diseases such as hereditary angioedema and surface-triggered thrombosis, and for developing treatments for thrombo-inflammatory disorders.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Factor XII Structure–Function RelationshipsShamanaev, AleksandrLitvak, MaximIvanov, IvanSrivastava, PriyankaSun, Mao-FuDickeson, S. KentKumar, SunilHe, Tracey Z.Gailani, DavidDOI:10.1055/s-0043-1769509Semin Thromb Hemost ; : -2023-06-05T06:36:21+01:00Seminars in Thrombosis and Hemostasis2023-06-05T06:36:21+01:00eFirst
    Review Article
    10.1055/s-0043-1769509http://dx.doi.org/10.1055/s-0043-1769509
    Inhibitors of Polyphosphate and Neutrophil Extracellular Trapshttp://dx.doi.org/10.1055/s-0043-1768936The contact pathway of blood clotting has received intense interest in recent years as studies have linked it to thrombosis, inflammation, and innate immunity. Because the contact pathway plays little to no role in normal hemostasis, it has emerged as a potential target for safer thromboprotection, relative to currently approved antithrombotic drugs which all target the final common pathway of blood clotting. Research since the mid-2000s has identified polyphosphate, DNA, and RNA as important triggers of the contact pathway with roles in thrombosis, although these molecules also modulate blood clotting and inflammation via mechanisms other than the contact pathway of the clotting cascade. The most significant source of extracellular DNA in many disease settings is in the form of neutrophil extracellular traps (NETs), which have been shown to contribute to incidence and severity of thrombosis. This review summarizes known roles of extracellular polyphosphate and nucleic acids in thrombosis, with an emphasis on novel agents under current development that target the prothrombotic activities of polyphosphate and NETs.Semin Thromb Hemost
    DOI: 10.1055/s-0043-1768936

    The contact pathway of blood clotting has received intense interest in recent years as studies have linked it to thrombosis, inflammation, and innate immunity. Because the contact pathway plays little to no role in normal hemostasis, it has emerged as a potential target for safer thromboprotection, relative to currently approved antithrombotic drugs which all target the final common pathway of blood clotting. Research since the mid-2000s has identified polyphosphate, DNA, and RNA as important triggers of the contact pathway with roles in thrombosis, although these molecules also modulate blood clotting and inflammation via mechanisms other than the contact pathway of the clotting cascade. The most significant source of extracellular DNA in many disease settings is in the form of neutrophil extracellular traps (NETs), which have been shown to contribute to incidence and severity of thrombosis. This review summarizes known roles of extracellular polyphosphate and nucleic acids in thrombosis, with an emphasis on novel agents under current development that target the prothrombotic activities of polyphosphate and NETs.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Inhibitors of Polyphosphate and Neutrophil Extracellular TrapsVappala, SreeparnaSmith, Stephanie A.Kizhakkedathu, Jayachandran N.Morrissey, James H.DOI:10.1055/s-0043-1768936Semin Thromb Hemost ; : -2023-05-16T06:50:55+01:00Seminars in Thrombosis and Hemostasis2023-05-16T06:50:55+01:00eFirst
    Review Article
    10.1055/s-0043-1768936http://dx.doi.org/10.1055/s-0043-1768936
    Plasma Kallikrein as a Forgotten Clotting Factorhttp://dx.doi.org/10.1055/s-0043-57034For decades, it was considered that plasma kallikrein's (PKa) sole function within the coagulation cascade is the activation of factor (F)XII. Until recently, the two key known activators of FIX within the coagulation cascade were activated FXI(a) and the tissue factor–FVII(a) complex. Simultaneously, and using independent experimental approaches, three groups identified a new branch of the coagulation cascade, whereby PKa can directly activate FIX. These key studies identified that (1) FIX or FIXa can bind with high affinity to either prekallikrein (PK) or PKa; (2) in human plasma, PKa can dose dependently trigger thrombin generation and clot formation independent of FXI; (3) in FXI knockout murine models treated with intrinsic pathway agonists, PKa activity results in increased formation of FIXa:AT complexes, indicating direct activation of FIX by PKa in vivo. These findings suggest that there is both a canonical (FXIa-dependent) and non-canonical (PKa-dependent) pathway of FIX activation. These three recent studies are described within this review, alongside historical data that hinted at the existence of this novel role of PKa as a coagulation clotting factor. The implications of direct PKa cleavage of FIX remain to be determined physiologically, pathophysiologically, and in the context of next-generation anticoagulants in development.Semin Thromb Hemost
    DOI: 10.1055/s-0043-57034

    For decades, it was considered that plasma kallikrein's (PKa) sole function within the coagulation cascade is the activation of factor (F)XII. Until recently, the two key known activators of FIX within the coagulation cascade were activated FXI(a) and the tissue factor–FVII(a) complex. Simultaneously, and using independent experimental approaches, three groups identified a new branch of the coagulation cascade, whereby PKa can directly activate FIX. These key studies identified that (1) FIX or FIXa can bind with high affinity to either prekallikrein (PK) or PKa; (2) in human plasma, PKa can dose dependently trigger thrombin generation and clot formation independent of FXI; (3) in FXI knockout murine models treated with intrinsic pathway agonists, PKa activity results in increased formation of FIXa:AT complexes, indicating direct activation of FIX by PKa in vivo. These findings suggest that there is both a canonical (FXIa-dependent) and non-canonical (PKa-dependent) pathway of FIX activation. These three recent studies are described within this review, alongside historical data that hinted at the existence of this novel role of PKa as a coagulation clotting factor. The implications of direct PKa cleavage of FIX remain to be determined physiologically, pathophysiologically, and in the context of next-generation anticoagulants in development.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Plasma Kallikrein as a Forgotten Clotting FactorKearney, Katherine J.Spronk, Henri M.H.Emsley, JonasKey, Nigel S.Philippou, HelenDOI:10.1055/s-0043-57034Semin Thromb Hemost ; : -2023-04-18T11:33:27+01:00Seminars in Thrombosis and Hemostasis2023-04-18T11:33:27+01:00eFirst
    Review Article
    10.1055/s-0043-57034http://dx.doi.org/10.1055/s-0043-57034
    Targeting the Contact Pathway of Coagulation for the Prevention and Management of Medical Device-Associated Thrombosishttp://dx.doi.org/10.1055/s-0043-57011Hemorrhage 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.Semin Thromb Hemost
    DOI: 10.1055/s-0043-57011

    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.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Targeting the Contact Pathway of Coagulation for the Prevention and Management of Medical Device-Associated ThrombosisGoel, AbhishekTathireddy, HarshaWang, Si-HanVu, Helen H.Puy, CristinaHinds, Monica T.Zonies, DavidMcCarty, Owen J.T.Shatzel, Joseph J.DOI:10.1055/s-0043-57011Semin Thromb Hemost ; : -2023-04-12T11:52:22+01:00Seminars in Thrombosis and Hemostasis2023-04-12T11:52:22+01:00eFirst
    Review Article
    10.1055/s-0043-57011http://dx.doi.org/10.1055/s-0043-57011
    Substrates, Cofactors, and Cellular Targets of Coagulation Factor XIahttp://dx.doi.org/10.1055/s-0043-1764469Coagulation 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.Semin Thromb Hemost
    DOI: 10.1055/s-0043-1764469

    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.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  Full text

    ]]>
    Substrates, Cofactors, and Cellular Targets of Coagulation Factor XIaLira, André L.Kohs, Tia C.L.Moellmer, Samantha A.Shatzel, Joseph J.McCarty, Owen J.T.Puy, CristinaDOI:10.1055/s-0043-1764469Semin Thromb Hemost ; : -2023-03-20T05:51:36+0100Seminars in Thrombosis and Hemostasis2023-03-20T05:51:36+0100eFirst
    Review Article
    10.1055/s-0043-1764469http://dx.doi.org/10.1055/s-0043-1764469
    Hereditary Angioedema: The Clinical Picture of Excessive Contact Activationhttp://dx.doi.org/10.1055/s-0042-1758820Hereditary angioedema is a rare, genetic disorder characterized by painful, debilitating and potentially life-threatening angioedema attacks in subcutaneous and submucosal tissue. While usually unpredictable, attacks can be provoked by a variety of triggers including physical injury and certain medication and are often preceded by prodromal symptoms. Hereditary angioedema has a profound influence on the patients' lives. The fundamental cause of hereditary angioedema in almost all patients is a mutation in the SERPING1 gene leading to a deficiency in C1-inhibitor. Subsequently, the contact activation cascade and kallikrein-kinin pathway are insufficiently inhibited, resulting in excessive bradykinin production triggering vascular leakage. While C1-inhibitor is an important regulator of the intrinsic coagulation pathway, fibrinolytic system and complement cascade, patients do not have an increased risk of coagulopathy, autoimmune conditions or immunodeficiency disorders. Hereditary angioedema is diagnosed based on C1-inhibitor level and function. Genetic analysis is only required in rare cases where hereditary angioedema with normal C1-inhibitor is found. In recent years, new, highly specific therapies have greatly improved disease control and angioedema-related quality of life. This article reviews the clinical picture of hereditary angioedema, the underlying pathophysiology, diagnostic process and currently available as well as investigational therapeutic options.Semin Thromb Hemost
    DOI: 10.1055/s-0042-1758820

    Hereditary angioedema is a rare, genetic disorder characterized by painful, debilitating and potentially life-threatening angioedema attacks in subcutaneous and submucosal tissue. While usually unpredictable, attacks can be provoked by a variety of triggers including physical injury and certain medication and are often preceded by prodromal symptoms. Hereditary angioedema has a profound influence on the patients' lives. The fundamental cause of hereditary angioedema in almost all patients is a mutation in the SERPING1 gene leading to a deficiency in C1-inhibitor. Subsequently, the contact activation cascade and kallikrein-kinin pathway are insufficiently inhibited, resulting in excessive bradykinin production triggering vascular leakage. While C1-inhibitor is an important regulator of the intrinsic coagulation pathway, fibrinolytic system and complement cascade, patients do not have an increased risk of coagulopathy, autoimmune conditions or immunodeficiency disorders. Hereditary angioedema is diagnosed based on C1-inhibitor level and function. Genetic analysis is only required in rare cases where hereditary angioedema with normal C1-inhibitor is found. In recent years, new, highly specific therapies have greatly improved disease control and angioedema-related quality of life. This article reviews the clinical picture of hereditary angioedema, the underlying pathophysiology, diagnostic process and currently available as well as investigational therapeutic options.
    [...]

    Thieme Medical Publishers, Inc. 333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

    Article in Thieme eJournals:
    Table of contents  |  Abstract  |  open access Full text

    ]]>
    Hereditary Angioedema: The Clinical Picture of Excessive Contact ActivationPetersen, Remy S.Fijen, Lauré M.Levi, MarcelCohn, Danny M.DOI:10.1055/s-0042-1758820Semin Thromb Hemost ; : -2022-11-23T06:37:31+0100Seminars in Thrombosis and Hemostasis2022-11-23T06:37:31+0100eFirst
    Review Article
    10.1055/s-0042-1758820http://dx.doi.org/10.1055/s-0042-1758820
    -