432 results on '"Douglas B, Cines"'
Search Results
2. Functional NMDA receptors are expressed by human pulmonary artery smooth muscle cells
- Author
-
Yi Na Dong, Fu-Chun Hsu, Cynthia J. Koziol-White, Victoria Stepanova, Joseph Jude, Andrei Gritsiuta, Ryan Rue, Rosalind Mott, Douglas A. Coulter, Reynold A. Panettieri, Vera P. Krymskaya, Hajime Takano, Elena A. Goncharova, Dmitry A. Goncharov, Douglas B. Cines, and David R. Lynch
- Subjects
Medicine ,Science - Abstract
Abstract N-methyl-d-aspartate (NMDA) receptors are widely expressed in the central nervous system. However, their presence and function at extraneuronal sites is less well characterized. In the present study, we examined the expression of NMDA receptor subunit mRNA and protein in human pulmonary artery (HPA) by quantitative polymerase chain reaction (PCR), immunohistochemistry and immunoblotting. We demonstrate that both GluN1 and GluN2 subunit mRNAs are expressed in HPA. In addition, GluN1 and GluN2 (A–D) subunit proteins are expressed by human pulmonary artery smooth muscle cells (HPASMCs) in vitro and in vivo. These subunits localize on the surface of HPASMCs and form functional ion channels as evidenced by whole-cell patch-clamp electrophysiology and reduced phenylephrine-induced contractile responsiveness of human pulmonary artery by the NMDA receptor antagonist MK801 under hypoxic condition. HPASMCs also express high levels of serine racemase and vesicular glutamate transporter 1, suggesting a potential source of endogenous agonists for NMDA receptor activation. Our findings show HPASMCs express functional NMDA receptors in line with their effect on pulmonary vasoconstriction, and thereby suggest a novel therapeutic target for pharmacological modulations in settings associated with pulmonary vascular dysfunction.
- Published
- 2021
- Full Text
- View/download PDF
3. The distinctive structure and composition of arterial and venous thrombi and pulmonary emboli
- Author
-
Irina N. Chernysh, Chandrasekaran Nagaswami, Sofia Kosolapova, Alina D. Peshkova, Adam Cuker, Douglas B. Cines, Carolyn L. Cambor, Rustem I. Litvinov, and John W. Weisel
- Subjects
Medicine ,Science - Abstract
Abstract Although arterial and venous thromboembolic disorders are among the most frequent causes of mortality and morbidity, there has been little description of how the composition of thrombi and emboli depends on their vascular origin and age. We quantified the structure and composition of arterial and venous thrombi and pulmonary emboli using high-resolution scanning electron microscopy. Arterial thrombi contained a surprisingly large amount of fibrin, in addition to platelets. The composition of pulmonary emboli mirrored the most distal part of venous thrombi from which they originated, which differed from the structure of the body and head of the same thrombi. All thrombi and emboli contained few biconcave red blood cells but many polyhedrocytes or related forms of compressed red blood cells, demonstrating that these structures are a signature of clot contraction in vivo. Polyhedrocytes and intermediate forms comprised the major constituents of venous thrombi and pulmonary emboli. The structures within all of the thrombi and emboli were very tightly packed, in contrast to clots formed in vitro. There are distinctive, reproducible differences among arterial and venous thrombi and emboli related to their origin, destination and duration, which may have clinical implications for the understanding and treatment of thrombotic disorders.
- Published
- 2020
- Full Text
- View/download PDF
4. American Society of Hematology 2019 guidelines for immune thrombocytopenia
- Author
-
Cindy Neunert, Deirdra R. Terrell, Donald M. Arnold, George Buchanan, Douglas B. Cines, Nichola Cooper, Adam Cuker, Jenny M. Despotovic, James N. George, Rachael F. Grace, Thomas Kühne, David J. Kuter, Wendy Lim, Keith R. McCrae, Barbara Pruitt, Hayley Shimanek, and Sara K. Vesely
- Subjects
Specialties of internal medicine ,RC581-951 - Abstract
Abstract: Background: Despite an increase in the number of therapies available to treat patients with immune thrombocytopenia (ITP), there are minimal data from randomized trials to assist physicians with the management of patients.Objective: These evidence-based guidelines of the American Society of Hematology (ASH) are intended to support patients, clinicians, and other health care professionals in their decisions about the management of ITP.Methods: In 2015, ASH formed a multidisciplinary guideline panel that included 8 adult clinical experts, 5 pediatric clinical experts, 2 methodologists with expertise in ITP, and 2 patient representatives. The panel was balanced to minimize potential bias from conflicts of interest. The panel reviewed the ASH 2011 guideline recommendations and prioritized questions. The panel used the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach, including evidence-to-decision frameworks, to appraise evidence (up to May 2017) and formulate recommendations.Results: The panel agreed on 21 recommendations covering management of ITP in adults and children with newly diagnosed, persistent, and chronic disease refractory to first-line therapy who have non–life-threatening bleeding. Management approaches included: observation, corticosteroids, IV immunoglobulin, anti-D immunoglobulin, rituximab, splenectomy, and thrombopoietin receptor agonists.Conclusions: There was a lack of evidence to support strong recommendations for various management approaches. In general, strategies that avoided medication side effects were favored. A large focus was placed on shared decision-making, especially with regard to second-line therapy. Future research should apply standard corticosteroid-dosing regimens, report patient-reported outcomes, and include cost-analysis evaluations.
- Published
- 2019
- Full Text
- View/download PDF
5. Vaccine-induced immune thrombotic thrombocytopenia
- Author
-
Douglas B. Cines and Andreas Greinacher
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Abstract
Within the first months of the COVID-19 vaccination campaign, previously healthy recipients who developed severe thrombosis (often cerebral and/or splanchnic vasculature) and thrombocytopenia typically after adenoviral vector-based vaccination were identified. Similarities between this syndrome, vaccine-induced immune thrombotic thrombocytopenia (VITT), and heparin-induced thrombocytopenia prompted recognition of the role of antiplatelet factor 4 (PF4) antibodies and management strategies based on IV immunoglobulin and nonheparin anticoagulants, which improved outcome. We update current understanding of VITT and potential involvement of anti-PF4 antibodies in thrombotic disorders.
- Published
- 2023
6. Modulation of ultralarge immune complexes in heparin–induced thrombocytopenia
- Author
-
Zheng Cai, Khalil Bdeir, Serge V. Yarovoi, Lubica Rauova, Gowthami M. Arepally, Sanjay Khandelwal, Jerome Rollin, Yves Gruel, Sergei Zaitsev, Mortimer Poncz, Mark I. Greene, and Douglas B. Cines
- Subjects
Hematology - Published
- 2023
7. Precision targeting of the plasminogen activator inhibitor‐1 mechanism increases efficacy of fibrinolytic therapy in empyema
- Author
-
Galina Florova, René A. Girard, Ali O. Azghani, Krishna Sarva, Ann Buchanan, Sophia Karandashova, Christian J. DeVera, Danna Morris, Mignote Chamiso, Kathleen Koenig, Douglas B. Cines, Steven Idell, and Andrey A. Komissarov
- Subjects
empyema ,fibrinolytic therapy ,molecular target ,plasminogen activator inhibitor‐1 ,single chain tissue plasminogen activator ,Physiology ,QP1-981 - Abstract
Abstract Plasminogen activator inhibitor‐1 (PAI‐1) is an endogenous irreversible inhibitor of tissue‐type (tPA) and urokinase (uPA) plasminogen activators. PAI‐1‐targeted fibrinolytic therapy (PAI‐1‐TFT) is designed to decrease the therapeutic dose of tPA and uPA, attenuating the risk of bleeding and other complications. Docking site peptide (DSP) mimics the part of the PAI‐1 reactive center loop that interacts with plasminogen activators, thereby affecting the PAI‐1 mechanism. We used DSP for PAI‐1‐TFT in two rabbit models: chemically induced pleural injury and Streptococcus pneumoniae induced empyema. These models feature different levels of inflammation and PAI‐1 expression. PAI‐1‐TFT with DSP (2.0 mg/kg) converted ineffective doses of single chain (sc) tPA (72.5 µg/kg) and scuPA (62.5 µg/kg) into effective ones in chemically induced pleural injury. DSP (2.0 mg/kg) was ineffective in S. pneumoniae empyema, where the level of PAI‐1 is an order of magnitude higher. DSP dose escalation to 8.0 mg/kg resulted in effective PAI‐1‐TFT with 0.25 mg/kg sctPA (1/8th of the effective dose of sctPA alone) in empyema. There was no increase in the efficacy of scuPA. PAI‐1‐TFT with DSP increases the efficacy of fibrinolytic therapy up to 8‐fold in chemically induced (sctPA and scuPA) and infectious (sctPA) pleural injury in rabbits. PAI‐1 is a valid molecular target in our model of S. pneumoniae empyema in rabbits, which closely recapitulates key characteristics of empyema in humans. Low‐dose PAI‐1‐TFT is a novel interventional strategy that offers the potential to improve fibrinolytic therapy for empyema in clinical practice.
- Published
- 2021
- Full Text
- View/download PDF
8. Opposing effects of HNP1 (α-defensin-1) on plasma cholesterol and atherogenesis.
- Author
-
Mohamed Higazi, Suhair Abdeen, Rami Abu-Fanne, Samuel N Heyman, Aseel Masarwy, Khalil Bdeir, Emad Maraga, Douglas B Cines, and Abd Al-Roof Higazi
- Subjects
Medicine ,Science - Abstract
Atherosclerosis, the predominant cause of death in well-resourced countries, may develop in the presence of plasma lipid levels within the normal range. Inflammation may contribute to lesion development in these individuals, but the underlying mechanisms are not well understood. Transgenic mice expressing α-def-1 released from activated neutrophils develop larger lipid and macrophage-rich lesions in the proximal aortae notwithstanding hypocholesterolemia caused by accelerated clearance of α-def-1/low-density lipoprotein (LDL) complexes from the plasma. The phenotype does not develop when the release of α-def-1 is prevented with colchicine. However, ApoE-/- mice crossed with α-def-1 mice or given exogenous α-def-1 develop smaller aortic lesions associated with reduced plasma cholesterol, suggesting a protective effect of accelerated LDL clearance. Experiments were performed to address this seeming paradox and to determine if α-def-1 might provide a means to lower cholesterol and thereby attenuate atherogenesis. We confirmed that exposing ApoE-/- mice to α-def-1 lowers total plasma cholesterol and decreases lesion size. However, lesion size was larger than in mice with total plasma cholesterol lowered to the same extent by inhibiting its adsorption or by ingesting a low-fat diet. Furthermore, α-def-1 levels correlated independently with lesion size in ApoE-/- mice. These studies show that α-def-1 has competing effects on atherogenesis. Although α-def-1 accelerates LDL clearance from plasma, it also stimulates deposition and retention of LDL in the vasculature, which may contribute to development of atherosclerosis in individuals with normal or even low plasma levels of cholesterol. Inhibiting α-def-1 may attenuate the impact of chronic inflammation on atherosclerotic vascular disease.
- Published
- 2020
- Full Text
- View/download PDF
9. Biocompatible coupling of therapeutic fusion proteins to human erythrocytes
- Author
-
Carlos H. Villa, Daniel C. Pan, Ian H. Johnston, Colin F. Greineder, Landis R. Walsh, Elizabeth D. Hood, Douglas B. Cines, Mortimer Poncz, Don L. Siegel, and Vladimir R. Muzykantov
- Subjects
Specialties of internal medicine ,RC581-951 - Abstract
Abstract: Carriage of drugs by red blood cells (RBCs) modulates pharmacokinetics, pharmacodynamics, and immunogenicity. However, optimal targets for attaching therapeutics to human RBCs and adverse effects have not been studied. We engineered nonhuman-primate single-chain antibody fragments (scFvs) directed to human RBCs and fused scFvs with human thrombomodulin (hTM) as a representative biotherapeutic cargo (hTM-scFv). Binding fusions to RBCs on band 3/glycophorin A (GPA; Wright b [Wrb] epitope) and RhCE (Rh17/Hr0 epitope) similarly endowed RBCs with hTM activity, but differed in their effects on RBC physiology. scFv and hTM-scFv targeted to band 3/GPA increased membrane rigidity and sensitized RBCs to hemolysis induced by mechanical stress, while reducing sensitivity to hypo-osmotic hemolysis. Similar properties were seen for other ligands bound to GPA and band 3 on human and murine RBCs. In contrast, binding of scFv or hTM-scFv to RhCE did not alter deformability or sensitivity to mechanical and osmotic stress at similar copy numbers bound per RBCs. Contrasting responses were also seen for immunoglobulin G antibodies against band 3, GPA, and RhCE. RBC-bound hTM-scFv generated activated protein C (APC) in the presence of thrombin, but RhCE-targeted hTM-scFv demonstrated greater APC generation per bound copy. Both Wrb- and RhCE-targeted fusion proteins inhibited fibrin deposition induced by tumor necrosis factor-α in an endothelialized microfluidic model using human whole blood. RhCE-bound hTM-scFv more effectively reduced platelet and leukocyte adhesion, whereas anti-Wrb scFv appeared to promote platelet adhesion. These data provide a translational framework for the development of engineered affinity ligands to safely couple therapeutics to human RBCs.
- Published
- 2018
- Full Text
- View/download PDF
10. Minimal role for the alternative pathway in complement activation by HIT immune complexes
- Author
-
Ayiesha P. Barnes, Sanjay Khandelwal, Simone Sartoretto, Sooho Myoung, Samuel J. Francis, Grace M. Lee, Lubica Rauova, Douglas B. Cines, Jon T. Skare, Charles E. Booth, Brandon L. Garcia, and Gowthami M. Arepally
- Subjects
Heparin ,Immunoglobulin G ,Esterases ,Humans ,Complement Factor D ,Antigen-Antibody Complex ,Complement System Proteins ,Hematology ,Thrombocytopenia ,Complement Activation ,Receptors, Complement - Abstract
Anti-platelet factor 4 (PF4)/heparin immune complexes that cause heparin-induced thrombocytopenia (HIT) activate complement via the classical pathway. Previous studies have shown that the alternative pathway of complement substantially amplifies the classical pathway of complement activation through the C3b feedback cycle.These studies sought to examine the contributions of the alternative pathway to complement activation by HIT antibodies.Using IgG monoclonal (KKO) and/or patient-derived HIT antibodies, we compared the effects of classical pathway (BBK32 and C1-esterase inhibitor [C1-INH]), alternative pathway (anti-factor B [fB] or factor D [fD] inhibitor) or combined classical and alternative pathway inhibition (soluble complement receptor 1 [sCR1]) in whole blood or plasma.Classical pathway inhibitors BBK32 and C1-INH and the combined classical/alternative pathway inhibitor sCR1 prevented KKO/HIT immune complex-induced complement activation, including release of C3 and C5 activation products, binding of immune complexes to B cells, and neutrophil activation. The alternative pathway inhibitors fB and fD, however, did not affect complement activation by KKO/HIT immune complexes. Similarly, alternative pathway inhibition had no effect on complement activation by unrelated immune complexes consisting of anti-dinitrophenyl (DNP) antibody and the multivalent DNP--keyhole limpet hemocyanin antigen.Collectively, these findings suggest the alternative pathway contributes little in support of complement activation by HIT immune complexes. Additional in vitro and in vivo studies are required to examine if this property is shared by most IgG-containing immune complexes or if predominance of the classic pathway is limited to immune complexes composed of multivalent antigens.
- Published
- 2022
11. ICAM-1–targeted thrombomodulin mitigates tissue factor–driven inflammatory thrombosis in a human endothelialized microfluidic model
- Author
-
Colin F. Greineder, Ian H. Johnston, Carlos H. Villa, Kandace Gollomp, Charles T. Esmon, Douglas B. Cines, Mortimer Poncz, and Vladimir R. Muzykantov
- Subjects
Specialties of internal medicine ,RC581-951 - Abstract
Abstract: Diverse human illnesses are characterized by loss or inactivation of endothelial thrombomodulin (TM), predisposing to microvascular inflammation, activation of coagulation, and tissue ischemia. Single-chain antibody fragment (scFv)/TM) fusion proteins, previously protective against end-organ injury in murine models of inflammation, are attractive candidates to treat inflammatory thrombosis. However, animal models have inherent differences in TM and coagulation biology, are limited in their ability to resolve and control endothelial biology, and do not allow in-depth testing of “humanized” scFv/TM fusion proteins, which are necessary for translation to the clinical domain. To address these challenges, we developed a human whole-blood, microfluidic model of inflammatory, tissue factor (TF)–driven coagulation that features a multichannel format for head-to-head comparison of therapeutic approaches. In this model, fibrin deposition, leukocyte adhesion, and platelet adhesion and aggregation showed a dose-dependent response to tumor necrosis factor-α activation and could be quantified via real-time microscopy. We used this model to compare hTM/R6.5, a humanized, intracellular adhesion molecule 1 (ICAM-1)–targeted scFv/TM biotherapeutic, to untargeted antithrombotic agents, including soluble human TM (shTM), anti–TF antibodies, and hirudin. The targeted hTM/R6.5 more effectively inhibited TF-driven coagulation in a protein C (PC)–dependent manner and demonstrated synergy with supplemental PC. These results support the translational prospects of ICAM-targeted scFv/TM and illustrate the utility of the microfluidic system as a platform to study humanized therapeutics at the interface of endothelium and whole blood under flow.
- Published
- 2017
- Full Text
- View/download PDF
12. The child with immune thrombocytopenia: to treat or not to treat, is that still the question?
- Author
-
Nichola Cooper and Douglas B. Cines
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2019
- Full Text
- View/download PDF
13. Prevention of thrombocytopenia and thrombosis in HIT using deglycosylated KKO: a novel therapeutic?
- Author
-
Amrita Sarkar, Sanjay Khandelwal, Gavin Koma, Hyunjun Kim, Yves Gruel, Jerome Rollin, Freda H Passam, Geoffrey D. Wool, Gowthami M. Arepally, Douglas B. Cines, Lubica Rauova, and Mortimer Poncz
- Subjects
Hematology - Abstract
Heparin-induced thrombocytopenia (HIT) is characterized by thrombocytopenia associated with a highly prothrombotic state due to the development of pathogenic antibodies that recognize human (h) platelet factor 4 (PF4) complexed with various polyanions. While non-heparin anticoagulants are the mainstay of care in HIT, subsequent bleeding may develop, and risk of new thromboembolic events remain. We had described a mouse IgGК2b antibody KKO that mimics the sentinel features of pathogenic HIT antibodies, including binding to the same neoepitope on hPF4:polyanion complexes. KKO, like HIT IgGs, both activates platelets through FcγRIIA and induces complement activation. We now asked whether Fc-modified KKO can be used as a novel therapeutic to prevent or treat HIT. Using the endoglycosidase EndoS, we created deglycosylated KKO (DGKKO). While DGKKO retained binding to PF4-polyanion complexes, it inhibited FcγRIIA-dependent activation of PF4-treated platelets triggered by unmodified KKO, 5B9 (another HIT-like monoclonal antibody), and IgGs isolated from HIT patients. DGKKO also decreased complement activation and deposition of C3c on platelets. Unlike the anticoagulant fondaparinux, injection of DGKKO into "HIT mice" lacking mouse PF4, but transgenic for hPF4 and FcγRIIA, prevented and reversed thrombocytopenia when injected before or after unmodified KKO, 5B9 or HIT IgG. DGKKO also reversed antibody-induced thrombus growth in HIT mice. In contrast, DGKKO was ineffective in preventing thrombosis by IgG from patients with the HIT-related anti-PF4 prothrombotic disorder, vaccine-induced immune thrombotic thrombocytopenia. Thus, DGKKO may represent a new class of therapeutics for targeted treatment of patients with HIT.
- Published
- 2023
14. Human neutrophil peptides 1-3 protect the murine urinary tract from uropathogenic
- Author
-
Jorge J, Canas, Dong, Liang, Vijay, Saxena, Jenaya, Hooks, Samuel W, Arregui, Hongyu, Gao, Yunlong, Liu, Danielle, Kish, Sarah C, Linn, Khalil, Bdeir, Douglas B, Cines, Robert L, Fairchild, John D, Spencer, Andrew L, Schwaderer, and David S, Hains
- Subjects
Mice ,alpha-Defensins ,DNA Copy Number Variations ,Pyelonephritis ,Genetic Loci ,Urinary Tract Infections ,Animals ,Humans ,Uropathogenic Escherichia coli ,Mice, Transgenic ,Urinary Tract ,Escherichia coli Infections - Abstract
Antimicrobial peptides (AMPs) are critical to the protection of the urinary tract of humans and other animals from pathogenic microbial invasion. AMPs rapidly destroy pathogens by disrupting microbial membranes and/or augmenting or inhibiting the host immune system through a variety of signaling pathways. We have previously demonstrated that alpha-defensins 1-3 (
- Published
- 2023
15. Polyphosphate/platelet factor 4 complexes can mediate heparin-independent platelet activation in heparin-induced thrombocytopenia
- Author
-
Douglas B. Cines, Serge V. Yarovoi, Sergei V. Zaitsev, Tatiana Lebedeva, Lubica Rauova, Mortimer Poncz, Gowthami M. Arepally, Sanjay Khandelwal, Victoria Stepanova, Ann H. Rux, Adam Cuker, Cecilia Guo, Linnette Mae Ocariza, Richard J. Travers, Stephanie A. Smith, Hugh Kim, James H. Morrissey, and Edward M. Conway
- Subjects
Specialties of internal medicine ,RC581-951 - Abstract
Abstract: Heparin-induced thrombocytopenia (HIT) is a thrombotic disorder initiated by antibodies to complexes between platelet factor 4 (PF4) and heparin. The risk of recurrent thromboembolism persists after heparin is cleared and platelet activation leading to release of PF4 has dissipated. We asked whether antigenic complexes between polyphosphates and PF4 released from activated platelets might intensify or sustain the prothrombotic phenotype of HIT. PF4 forms stable, ultralarge complexes with polyphosphates of various sizes, including those released from platelets, which are recognized by the HIT-like monoclonal KKO, an immunoglobulin G2bκ monoclonal heparin/PF4 binding antibody, and by human HIT antibodies. KKO helps to protect PF4/polyphosphate complexes from degradation by phosphatases. Complement is activated when HIT antibodies bind to PF4/polyphosphate complexes and PF4 reverses the inhibition of complement by polyphosphates. Polyphosphates and PF4 are stored primarily in separate granules in resting platelets, but they colocalize when the cells are activated. Platelets activated by subaggregating doses of thrombin receptor activating peptide release polyphosphates and PF4, which form antigenic complexes that allow KKO to further activate platelets in the absence of heparin and exogenous PF4. These studies suggest that thrombin- or immune complex–mediated release of endogenous antigenic PF4/polyphosphate complexes from platelets may augment the prothrombotic risk of HIT and perpetuate the risk of thrombosis after heparin has been discontinued.
- Published
- 2016
- Full Text
- View/download PDF
16. Neutrophil extracellular trap stabilization by platelet factor 4 reduces thrombogenicity and endothelial cell injury
- Author
-
Anh T. P. Ngo, Amrita Sarkar, Irene Yarovoi, Nate D. Levine, Veronica Bochenek, Guohua Zhao, Lubica Rauova, M. Anna Kowalska, Kaitlyn Eckart, Nilam S. Mangalmurti, Ann Rux, Douglas B. Cines, Mortimer Poncz, and Kandace Gollomp
- Subjects
Article - Abstract
Neutrophil extracellular traps (NETs) are abundant in sepsis, and proposed NET-directed therapies in sepsis prevent their formation or accelerate degradation. Yet NETs are important for microbial entrapment, as NET digestion liberates pathogens and NET degradation products (NDPs) that deleteriously promote thrombosis and endothelial cell injury. We proposed an alternative strategy of NET-stabilization with the chemokine, platelet factor 4 (PF4, CXCL4), which we have shown enhances NET-mediated microbial entrapment. We now show that NET compaction by PF4 reduces their thrombogenicity. In vitro, we quantified plasma thrombin and fibrin generation by intact or degraded NETs and cell-free (cf) DNA fragments, and found that digested NETs and short DNA fragments were more thrombogenic than intact NETs and high molecular weight genomic DNA, respectively. PF4 reduced the thrombogenicity of digested NETs and DNA by interfering, in part, with contact pathway activation. In endothelial cell culture studies, short DNA fragments promoted von Willebrand factor release and tissue factor expression via a toll-like receptor 9-dependent mechanism. PF4 blocked these effects.Cxcl4-/-mice infused with cfDNA exhibited higher plasma thrombin anti-thrombin (TAT) levels compared to wild-type controls. Following challenge with bacterial lipopolysaccharide,Cxcl4-/-mice had similar elevations in plasma TAT and cfDNA, effects prevented by PF4 infusion. Thus, NET-stabilization by PF4 prevents the release of short fragments of cfDNA, limiting the activation of the contact coagulation pathway and reducing endothelial injury. These results support our hypothesis that NET-stabilization reduces pathologic sequelae in sepsis, an observation of potential clinical benefit.HIGHLIGHTSIn contrast to intact NETs, degraded NETs and cfDNA are prothrombotic and injure the endothelium.PF4 reduces the ability of degraded NETs and cfDNA to promote thrombosis and injure the endothelium.
- Published
- 2023
17. Divergent impacts of tocilizumab and colchicine in COVID‐19‐associated coagulopathy: the role of alpha‐defensins
- Author
-
Mohamed Higazi, Douglas B. Cines, Suhair Abdeen, Abd Al-Roof Higazi, Rami Abu-Fanne, Emad Maraga, Khalil Bdeir, and Samuel N. Heyman
- Subjects
Male ,alpha-Defensins ,Neutrophils ,Anti-Inflammatory Agents ,Alpha (ethology) ,Inflammation ,Pharmacology ,Antibodies, Monoclonal, Humanized ,Fibrin Fibrinogen Degradation Products ,chemistry.chemical_compound ,Tocilizumab ,Correspondence ,Coagulopathy ,medicine ,Humans ,Colchicine ,Receptor ,Blood Coagulation ,thrombosis ,Aged ,Interleukin-6 ,business.industry ,COVID-19 ,Hematology ,Middle Aged ,medicine.disease ,In vitro ,COVID-19 Drug Treatment ,chemistry ,inflammation ,Female ,medicine.symptom ,Cytokine Release Syndrome ,business ,Cytokine storm - Abstract
Summary Patients who are severely affected by coronavirus disease 2019 (COVID‐19) may develop a delayed onset ‘cytokine storm’, which includes an increase in interleukin‐6 (IL‐6). This may be followed by a pro‐thrombotic state and increased D‐dimers. It was anticipated that tocilizumab (TCZ), an anti‐IL‐6 receptor monoclonal antibody, would mitigate inflammation and coagulation in patients with COVID‐19. However, clinical trials with TCZ have recorded an increase in D‐dimer levels. In contrast to TCZ, colchicine reduced D‐dimer levels in patients with COVID‐19. To understand how the two anti‐inflammatory agents have diverse effects on D‐dimer levels, we present data from two clinical trials that we performed. In the first trial, TCZ was administered (8 mg/kg) to patients who had a positive polymerase chain reaction test for COVID‐19. In the second trial, colchicine was given (0·5 mg twice a day). We found that TCZ significantly increased IL‐6, α‐Defensin (α‐Def), a pro‐thrombotic peptide, and D‐dimers. In contrast, treatment with colchicine reduced α‐Def and Di‐dimer levels. In vitro studies show that IL‐6 stimulated the release of α‐Def from human neutrophils but in contrast to colchicine, TCZ did not inhibit the stimulatory effect of IL‐6; raising the possibility that the increase in IL‐6 in patients with COVID‐19 treated with TCZ triggers the release of α‐Def, which promotes pro‐thrombotic events reflected in an increase in D‐dimer levels.
- Published
- 2021
18. Prevention of thrombocytopenia and thrombosis in heparin-induced thrombocytopenia (HIT) using deglycosylated KKO: A novel therapeutic?
- Author
-
Amrita Sarkar, Sanjay Khandelwal, Hyunjun Kim, Yves Gruel, Jerome Rollin, Geoffrey D. Wool, Gowthami M. Arepally, Douglas B. Cines, Lubica Rauova, and Mortimer Poncz
- Abstract
Heparin-induced thrombocytopenia (HIT) is characterized by mild thrombocytopenia associated with a highly prothrombotic state due to the development of pathogenic antibodies that recognize human (h) platelet factor 4 (PF4) complexed with various polyanions. While non-heparin anticoagulants and intravenous immunoglobulin (IVIG) are the mainstay of care, bleeding may develop, and risk of new thromboembolic events remain. We had described a mouse IgGκ2b antibody KKO that mimics the sentinel features of pathogenic HIT antibodies, including binding to the same neoepitope on hPF4:polyanion complexes. KKO, like HIT IgGs, activates platelets through FcγRIIA and induces complement activation. We now asked whether Fc-modified KKO can be used as a novel therapeutic to prevent or treat HIT. Using the endoglycosidase EndoS, we created deglycosylated KKO (DGKKO). DGKKO bound to PF4-polyanion complexes, and blocked FcγRIIA-dependent activation of PF4 treated platelets by KKO, 5B9 (another HIT-like monoclonal antibody), and isolated IgGs from HIT patients. DGKKO also decreased complement activation and deposition of C3c on platelets. Injection of DGKKO into “HIT mice” lacking mouse PF4, but transgenic for hPF4 and FcγRIIA, prevented and reversed thrombocytopenia when injected before or after KKO, 5B9 or HIT IgG, respectively, in a microfluidic system. DGKKO reversed antibody-induced thrombus growth in HIT mice. In contrast, DGKKO was ineffective in preventing thrombosis by IgG from a patient with the HIT-related disorder, vaccine-induced immune thrombotic thrombocytopenia. Thus, DGKKO may represent a new class of therapeutics for targeted treatment of patients with HIT.Key PointsDeglycosylated (DG) KKO can reverse thrombocytopenia in a HIT murine model.DGKKO can prevent/reverse thrombosisin vitroand in a HIT murine model.
- Published
- 2022
19. Human neutrophil peptides 1-3 protect the murine urinary tract from uropathogenic Escherichia coli challenge
- Author
-
Jorge J. Canas, Dong Liang, Vijay Saxena, Jenaya Hooks, Samuel W. Arregui, Hongyu Gao, Yunlong Liu, Danielle Kish, Sarah C. Linn, Khalil Bdeir, Douglas B. Cines, Robert L. Fairchild, John D. Spencer, Andrew L. Schwaderer, and David S. Hains
- Subjects
Multidisciplinary - Abstract
Antimicrobial peptides (AMPs) are critical to the protection of the urinary tract of humans and other animals from pathogenic microbial invasion. AMPs rapidly destroy pathogens by disrupting microbial membranes and/or augmenting or inhibiting the host immune system through a variety of signaling pathways. We have previously demonstrated that alpha-defensins 1-3 ( DEFA1A3 ) are AMPs expressed in the epithelial cells of the human kidney collecting duct in response to uropathogens. We also demonstrated that DNA copy number variations in the DEFA1A3 locus are associated with UTI and pyelonephritis risk. Because DEFA1A3 is not expressed in mice, we utilized human DEFA1A3 gene transgenic mice ( DEFA 4/4 ) to further elucidate the biological relevance of this locus in the murine urinary tract. We demonstrate that the kidney transcriptional and translational expression pattern is similar in humans and the human gene transgenic mouse upon uropathogenic Escherichia coli (UPEC) stimulus in vitro and in vivo. We also demonstrate transgenic human DEFA 4/4 gene mice are protected from UTI and pyelonephritis under various UPEC challenges. This study serves as the foundation to start the exploration of manipulating the DEFA1A3 locus and alpha-defensins 1-3 expression as a potential therapeutic target for UTIs and other infectious diseases.
- Published
- 2022
20. Safety and efficacy of romiplostim in splenectomized and nonsplenectomized patients with primary immune thrombocytopenia
- Author
-
Douglas B. Cines, Jeffrey Wasser, Francesco Rodeghiero, Beng H. Chong, Michael Steurer, Drew Provan, Roger Lyons, Jaime Garcia-Chavez, Nancy Carpenter, Xuena Wang, and Melissa Eisen
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Primary immune thrombocytopenia is an autoimmune disorder characterized by increased platelet destruction and insufficient platelet production without another identified underlying disorder. Splenectomy may alter responsiveness to treatment and/or increase the risk of thrombosis, infection, and pulmonary hypertension. The analysis herein evaluated the safety and efficacy of the thrombopoietin receptor agonist romiplostim in splenectomized and nonsplenectomized adults with primary immune thrombocytopenia. Data were pooled across 13 completed clinical studies in adults with immune thrombocytopenia from 2002–2014. Adverse event rates were adjusted for time of exposure. Results were considered different when 95% confidence intervals were non-overlapping. Safety was analyzed for 1111 patients (395 splenectomized; 716 nonsplenectomized) who received romiplostim or control (placebo or standard of care). At baseline, splenectomized patients had a longer median duration of immune thrombocytopenia and a lower median platelet count, as well as a higher proportion with >3 prior immune thrombocytopenia treatments versus nonsplenectomized patients. In each treatment group, splenectomized patients used rescue medications more often than nonsplenectomized patients. Platelet response rates (≥50×109/L) for romiplostim were 82% (310/376) for splenectomized and 91% (592/648) for nonsplenectomized patients (P
- Published
- 2017
- Full Text
- View/download PDF
21. Functional NMDA receptors are expressed by human pulmonary artery smooth muscle cells
- Author
-
Douglas B. Cines, Yi Na Dong, Joseph A. Jude, Andrei I. Gritsiuta, Victoria Stepanova, Ryan Rue, Douglas A. Coulter, Fu-Chun Hsu, David A. Lynch, Rosalind E. Mott, Elena A. Goncharova, Hajime Takano, Vera P. Krymskaya, Cynthia J. Koziol-White, Reynold A. Panettieri, and Dmitry A. Goncharov
- Subjects
0301 basic medicine ,Vesicular glutamate transporter 1 ,Inbred C57BL ,Cardiovascular ,Muscle, Smooth, Vascular ,Mice ,0302 clinical medicine ,Smooth Muscle ,Hypoxic pulmonary vasoconstriction ,Receptors ,Receptor ,Lung ,Cells, Cultured ,Cultured ,Multidisciplinary ,Ligand-gated ion channels ,biology ,Chemistry ,Respiration ,Cell biology ,Muscle ,NMDA receptor ,Medicine ,Smooth ,N-Methyl-D-Aspartate ,Cells ,Protein subunit ,Science ,Myocytes, Smooth Muscle ,Pulmonary Artery ,Receptors, N-Methyl-D-Aspartate ,Article ,03 medical and health sciences ,In vivo ,Vascular ,Genetics ,Animals ,Humans ,Ion channel ,Myocytes ,Neurosciences ,Mice, Inbred C57BL ,Protein Subunits ,030104 developmental biology ,Vasoconstriction ,Serine racemase ,biology.protein ,030217 neurology & neurosurgery - Abstract
N-methyl-d-aspartate (NMDA) receptors are widely expressed in the central nervous system. However, their presence and function at extraneuronal sites is less well characterized. In the present study, we examined the expression of NMDA receptor subunit mRNA and protein in human pulmonary artery (HPA) by quantitative polymerase chain reaction (PCR), immunohistochemistry and immunoblotting. We demonstrate that both GluN1 and GluN2 subunit mRNAs are expressed in HPA. In addition, GluN1 and GluN2 (A–D) subunit proteins are expressed by human pulmonary artery smooth muscle cells (HPASMCs) in vitro and in vivo. These subunits localize on the surface of HPASMCs and form functional ion channels as evidenced by whole-cell patch-clamp electrophysiology and reduced phenylephrine-induced contractile responsiveness of human pulmonary artery by the NMDA receptor antagonist MK801 under hypoxic condition. HPASMCs also express high levels of serine racemase and vesicular glutamate transporter 1, suggesting a potential source of endogenous agonists for NMDA receptor activation. Our findings show HPASMCs express functional NMDA receptors in line with their effect on pulmonary vasoconstriction, and thereby suggest a novel therapeutic target for pharmacological modulations in settings associated with pulmonary vascular dysfunction.
- Published
- 2021
22. Aggregation of Enveloped Viruses By Platelet Factor 4 (PF4) and Neutrophil Extracellular Traps (NETs): Implications for the Anti-Viral Properties of NETs
- Author
-
Kandace Gollomp, Irina Chernysh, Anh T.P. Ngo, Nathan Levine, Sergey Zaytsev, Veronica Bochenek, Lubica Rauova, Nicholas Parenti, Susan R Weiss, John W. Weisel, Douglas B. Cines, and Mortimer Poncz
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
23. Neutrophil Extracellular Traps (NETs) Thrombogenicity and Endothelial Cell Toxicity Are Ameliorated By Platelet Factor 4 (PF4): Implications to the Treatment of Sepsis
- Author
-
Anh T.P. Ngo, Amrita Sarkar, Nathan Levine, Veronica Bochenek, Guohua Zhao, Lubica Rauova, M. Anna Kowalska, Kaitlyn Eckart, Nilam S. Mangalmurti, Ann Rux, Douglas B. Cines, Mortimer Poncz, and Kandace Gollomp
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
24. Complement Shields Fc Regions on HIT Immune Complexes and Prevents Recognition By Cellular Fcγ Receptors
- Author
-
Hailey Harris, Sanjay Khandelwal, Lubica Rauova, John Lambris, Brandon Garcia, Douglas B. Cines, and Gowthami M. Arepally
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
25. Fusion of uPA to Anti-aIIbb3 Scfv Facilitates Its Uptake By in Vitro-Generated CD34+ Megakaryocytes (MKs) to Promote Targeted Fibrinolysis
- Author
-
Sergey Zaytsev, Hyunsook Ahn, M. Anna Kowalska, Rodney Camire, Douglas B. Cines, Mortimer Poncz, and Victoria Stepanova
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
26. Modulation of Ultralarge Immune Complexes in Heparin-Induced Thrombocytopenia
- Author
-
Lubica Rauova, Khalil Bdeir, Zheng Cai, Serge Yarovoi, Gowthami M. Arepally, Sanjay Khandelwal, Mortimer Poncz, Mark Greene, and Douglas B. Cines
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
27. Platelets get particular
- Author
-
Lubica Rauova and Douglas B. Cines
- Subjects
Blood Platelets ,ChAdOx1 nCoV-19 ,Immunology ,Immunity ,Humans ,Cell Biology ,Hematology ,Leukopenia ,Biochemistry ,Thrombocytopenia - Published
- 2022
28. Pathogenesis of heparin-induced thrombocytopenia
- Author
-
Douglas B. Cines and Gowthami M. Arepally
- Subjects
0301 basic medicine ,Article ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Physiology (medical) ,Heparin-induced thrombocytopenia ,medicine ,Humans ,Platelet ,biology ,Heparin ,business.industry ,Biochemistry (medical) ,Public Health, Environmental and Occupational Health ,Anticoagulants ,General Medicine ,Neutrophil extracellular traps ,medicine.disease ,Thrombocytopenia ,030104 developmental biology ,030220 oncology & carcinogenesis ,Immunology ,biology.protein ,Antibody ,business ,Platelet factor 4 ,medicine.drug - Abstract
There are currently no effective substitutes for high intensity therapy with unfractionated heparin (UFH) for cardiovascular procedures based on its rapid onset of action, ease of monitoring and reversibility. The continued use of UFH in these and other settings requires vigilance for its most serious non-hemorrhagic complication, heparin induced thrombocytopenia (HIT). HIT is an immune prothrombotic disorder caused by antibodies that recognize complexes between platelet factor 4 (PF4) and polyanions such as heparin (H). The pathogenicity of anti-PF4/H antibodies is likely due to the formation of immune complexes that initiate intense procoagulant responses by vascular and hematopoietic cells that lead to the generation of platelet microparticles, monocyte and endothelial cell procoagulant activity, and neutrophil extracellular traps (NETs), among other outcomes. The development of anti-PF4/H antibodies after exposure to UFH greatly exceeds the incidence of clinical disease, but the biochemical features that distinguish pathogenic from non-pathogenic antibodies have not been identified. Diagnosis relies on pretest clinical probability, screening for anti-PF4/H antibodies and documentation of their platelet activating capacity. However, both clinical algorithms and test modalities have limited predictive values making diagnosis and management challenging. Given the unacceptable rates of recurrent thromboembolism and bleeding associated with current therapies, there is an unmet need for novel rational non-anticoagulant therapeutics based on the pathogenesis of HIT. We will review recent developments in our understanding of the pathogenesis of HIT and its implications for future approaches to diagnosis and management.
- Published
- 2020
29. SARS-CoV-2 vaccination and ITP in patients with de novo or preexisting ITP
- Author
-
James B. Bussel, David J. Kuter, Alexandra Kruse, Andrew D. Leavitt, Douglas B. Cines, Howard A. Liebman, Jennifer DiRaimo, Terry Gernsheimer, Jecko Thachil, Caroline Kruse, Marina Beltrami Moreira, Adam Cuker, Craig M. Kessler, Michael D. Tarantino, Ashley Ray, Eun-Ju Lee, Hanny Al-Samkari, Adrian C. Newland, and Alfred Ian Lee
- Subjects
Blood Platelets ,Male ,medicine.medical_specialty ,COVID-19 Vaccines ,Exacerbation ,Platelet disorder ,medicine.medical_treatment ,Immunology ,Splenectomy ,Biochemistry ,immune system diseases ,Risk Factors ,hemic and lymphatic diseases ,Internal medicine ,Medicine ,Humans ,Platelet ,Adverse effect ,Aged ,Retrospective Studies ,Aged, 80 and over ,Purpura, Thrombocytopenic, Idiopathic ,biology ,business.industry ,SARS-CoV-2 ,COVID-19 ,Retrospective cohort study ,Cell Biology ,Hematology ,Middle Aged ,United Kingdom ,Vaccination ,biology.protein ,Female ,Antibody ,business - Abstract
Cases of de novo immune thrombocytopenia (ITP), including a fatality, following SARS-CoV-2 vaccination in previously healthy recipients led to studying its impact in preexisting ITP. In this study, 4 data sources were analyzed: the Vaccine Adverse Events Reporting System (VAERS) for cases of de novo ITP; a 10-center retrospective study of adults with preexisting ITP receiving SARS-CoV-2 vaccination; and surveys distributed by the Platelet Disorder Support Association (PDSA) and the United Kingdom (UK) ITP Support Association. Seventy-seven de novo ITP cases were identified in VAERS, presenting with median platelet count of 3 [1-9] ×109/L approximately 1 week postvaccination. Of 28 patients with available data, 26 responded to treatment with corticosteroids and/or intravenous immunoglobulin (IVIG), and/or platelet transfusions. Among 117 patients with preexisting ITP who received a SARS-CoV-2 vaccine, 19 experienced an ITP exacerbation (any of: ≥50% decline in platelet count, nadir platelet count 20% decrease from baseline, and/or use of rescue therapy) following the first dose and 14 of 70 after a second dose. Splenectomized persons and those who received 5 or more prior lines of therapy were at highest risk of ITP exacerbation. Fifteen patients received and responded to rescue treatment. In surveys of both 57 PDSA and 43 UK patients with ITP, prior splenectomy was associated with worsened thrombocytopenia. ITP may worsen in preexisting ITP or be identified de novo post–SARS-CoV2 vaccination; both situations responded well to treatment. Proactive monitoring of patients with known ITP, especially those postsplenectomy and with more refractory disease, is indicated.
- Published
- 2021
30. Erythrocytes identify complement activation in patients with COVID-19
- Author
-
Ariel R. Weisman, Caroline A. G. Ittner, Ann H. Rux, Nuala J. Meyer, S. Murphy, Wen-Chao Song, John D. Lambris, Nilam S. Mangalmurti, Michael R. Betts, Leticia Kuri-Cervantes, Douglas B. Cines, John P. Reilly, E. John Wherry, M. Betina Pampena, and L. K. Metthew Lam
- Subjects
Pulmonary and Respiratory Medicine ,Rapid Report ,Physiology ,business.industry ,Immune adherence ,Cell Biology ,Complement receptor ,Disease ,Complement C4b ,medicine.disease ,Complement system ,Complement (complexity) ,Sepsis ,Immune system ,Physiology (medical) ,Immunology ,medicine ,business - Abstract
COVID-19, the disease caused by the SARS-CoV-2 virus, can progress to multi-organ failure characterized by respiratory insufficiency, arrhythmias, thromboembolic complications and shock. The mortality of patients hospitalized with COVID-19 is unacceptably high and new strategies are urgently needed to rapidly identify and treat patients at risk for organ failure. Clinical epidemiologic studies demonstrate that vulnerability to organ failure is greatest after viral clearance from the upper airway, which suggests that dysregulation of the host immune response is a critical mediator of clinical deterioration and death. Autopsy and pre-clinical evidence implicate aberrant complement activation in endothelial injury and organ failure. A potential therapeutic strategy warranting investigation is to inhibit complement, with case reports of successful treatment of COVID-19 with inhibitors of complement. However, this approach requires careful balance between the host protective and potential injurious effects of complement activation, and biomarkers to identify the optimal timing and candidates for therapy are lacking. Here we report the presence of complement activation products on circulating erythrocytes from hospitalized COVID-19 patients using flow cytometry. These findings suggest that novel erythrocyte-based diagnostics provide a method to identify patients with dysregulated complement activation.
- Published
- 2021
31. Human neutrophil peptide‐1 inhibits thrombus formation under arterial flow via its terminal free cysteine thiols
- Author
-
Mohammad S. Abdelgawwad, Jenny K. McDaniel, Douglas B. Cines, Khalil Bdeir, Matthew B. Renfrow, Audra A. Hargett, Wenjing Cao, and X. Long Zheng
- Subjects
Blood Platelets ,alpha-Defensins ,Platelet Aggregation ,Endothelium ,ADAMTS13 Protein ,030204 cardiovascular system & hematology ,Article ,Cell Line ,03 medical and health sciences ,Platelet Adhesiveness ,0302 clinical medicine ,Von Willebrand factor ,von Willebrand Factor ,medicine ,Animals ,Humans ,Platelet ,Cysteine ,Sulfhydryl Compounds ,Blood Coagulation ,Mice, Knockout ,biology ,Chemistry ,Endothelial Cells ,Thrombosis ,Biological activity ,Hematology ,Adhesion ,Cell biology ,Endothelial stem cell ,medicine.anatomical_structure ,Regional Blood Flow ,Hemostasis ,biology.protein ,Tumor necrosis factor alpha ,Collagen - Abstract
Essentials Biological activity of human neutrophil peptide (HNP)-1 in hemostasis under physiological conditions is not fully understood. HNP-1 inhibits the adhesion/aggregation of murine platelets on a fibrillar collagen surface or an activated endothelial cell surface under flow. The anti-adhesion activity appears to depend on the terminal free thiols of HNP-1, which may inhibit VWF-VWF lateral associations. Our results suggest a protective role and potential novel therapeutic use of HNP-1 for arterial thrombosis. SUMMARY: Background Human neutrophil peptides (HNPs), also known as α-defensins, are released from degranulated neutrophils and play an important role in innate immunity. However, their biological roles in hemostasis under flow are not fully explored. Objective This study aims to determine the role of HNP-1 on platelet adhesion and aggregation on a collagen surface or ultra large von Willebrand factor (ULVWF) on endothelium under flow and elucidate the structural elements required for its activity. Methods Anticoagulated whole blood from wild-type or Adamts13-/- mice was incubated with a fluorescein-conjugated anti-human CD41 in the presence of increasing concentrations of a synthetic HNP-1 and perfused over a collagen surface or a tumor necrosis factor (TNF)-α activated murine endothelial cell surface under arterial flow. The rate of accumulation and the final surface coverage of fluoresceinated murine platelets or the rate of forming platelet-decorated ULVWF strings were determined using the BioFlux microfluidic system. Results HNP-1 inhibited the rate and final coverage of fluorescein-labeled murine platelets on a fibrillar collagen surface under flow (100 dyne/cm2 ) in a concentration-dependent manner and the anti-adhesive activity of HNP-1 depended on its terminal free cysteine thiols. HNP-1 (20 μM) also dramatically inhibited the formation of platelets-decorated ULVWF strings on TNF-α activated murine endothelial surface under arterial flow. Conclusions Our results demonstrate for the first time an antiplatelet adhesion or antithrombotic activity of HNP-1; this activity depends on its terminal free thiols, likely affecting VWF-VWF lateral associations. These findings may suggest a potential novel therapeutic strategy for arterial thrombosis.
- Published
- 2019
32. Neutrophil α-defensins promote thrombosis in vivo by altering fibrin formation, structure, and stability
- Author
-
Chandrasekaran Nagaswami, Khalil Bdeir, Alexander R. Mukhitov, Mohamed Higazi, Abd Al-Roof Higazi, Emad Maraga, John W. Weisel, Rustem I. Litvinov, Suhair Abdeen, Rami Abu-Fanne, Douglas B. Cines, and Victoria Stepanova
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Immunology ,Inflammation ,030204 cardiovascular system & hematology ,Biochemistry ,Inferior vena cava ,Fibrin ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Fibrinolysis ,medicine ,biology ,Chemistry ,Cell Biology ,Hematology ,Heparin ,Cell biology ,030104 developmental biology ,medicine.vein ,Coagulation ,Neutrophil degranulation ,biology.protein ,medicine.symptom ,medicine.drug - Abstract
Inflammation and thrombosis are integrated, mutually reinforcing processes, but the interregulatory mechanisms are incompletely defined. Here, we examined the contribution of α-defensins (α-defs), antimicrobial proteins released from activated human neutrophils, on clot formation in vitro and in vivo. Activation of the intrinsic pathway of coagulation stimulates release of α-defs from neutrophils. α-Defs accelerate fibrin polymerization, increase fiber density and branching, incorporate into nascent fibrin clots, and impede fibrinolysis in vitro. Transgenic mice (Def++) expressing human α-Def-1 developed larger, occlusive, neutrophil-rich clots after partial inferior vena cava (IVC) ligation than those that formed in wild-type (WT) mice. IVC thrombi extracted from Def++ mice were composed of a fibrin meshwork that was denser and contained a higher proportion of tightly packed compressed polyhedral erythrocytes than those that developed in WT mice. Def++ mice were resistant to thromboprophylaxis with heparin. Inhibiting activation of the intrinsic pathway of coagulation, bone marrow transplantation from WT mice or provision of colchicine to Def++ mice to inhibit neutrophil degranulation decreased plasma levels of α-defs, caused a phenotypic reversion characterized by smaller thrombi comparable to those formed in WT mice, and restored responsiveness to heparin. These data identify α-defs as a potentially important and tractable link between innate immunity and thrombosis.
- Published
- 2019
33. Precision targeting of the plasminogen activator inhibitor‐1 mechanism increases efficacy of fibrinolytic therapy in empyema
- Author
-
Steven Idell, Andrey A. Komissarov, Kathleen Koenig, Ann Buchanan, Douglas B. Cines, Ali Azghani, Krishna Sarva, Sophia Karandashova, Mignote Chamiso, D.E. Morris, R. Girard, Galina Florova, and Christian J. DeVera
- Subjects
Physiology ,Inflammation ,Endogeny ,030204 cardiovascular system & hematology ,Pharmacology ,medicine.disease_cause ,molecular target ,Plasminogen Activators ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Therapeutic index ,single chain tissue plasminogen activator ,Physiology (medical) ,Plasminogen Activator Inhibitor 1 ,Streptococcus pneumoniae ,medicine ,Animals ,QP1-981 ,Thrombolytic Therapy ,Reactive center ,Urokinase ,Binding Sites ,business.industry ,Original Articles ,medicine.disease ,Effective dose (pharmacology) ,Empyema ,respiratory tract diseases ,chemistry ,Plasminogen activator inhibitor-1 ,empyema ,plasminogen activator inhibitor‐1 ,Female ,Original Article ,Rabbits ,medicine.symptom ,fibrinolytic therapy ,business ,Oligopeptides ,Plasminogen activator ,030217 neurology & neurosurgery ,Protein Binding ,medicine.drug - Abstract
Plasminogen activator inhibitor‐1 (PAI‐1) is an endogenous irreversible inhibitor of tissue‐type (tPA) and urokinase (uPA) plasminogen activators. PAI‐1‐targeted fibrinolytic therapy (PAI‐1‐TFT) is designed to decrease the therapeutic dose of tPA and uPA, attenuating the risk of bleeding and other complications. Docking site peptide (DSP) mimics the part of the PAI‐1 reactive center loop that interacts with plasminogen activators, thereby affecting the PAI‐1 mechanism. We used DSP for PAI‐1‐TFT in two rabbit models: chemically induced pleural injury and Streptococcus pneumoniae induced empyema. These models feature different levels of inflammation and PAI‐1 expression. PAI‐1‐TFT with DSP (2.0 mg/kg) converted ineffective doses of single chain (sc) tPA (72.5 µg/kg) and scuPA (62.5 µg/kg) into effective ones in chemically induced pleural injury. DSP (2.0 mg/kg) was ineffective in S. pneumoniae empyema, where the level of PAI‐1 is an order of magnitude higher. DSP dose escalation to 8.0 mg/kg resulted in effective PAI‐1‐TFT with 0.25 mg/kg sctPA (1/8th of the effective dose of sctPA alone) in empyema. There was no increase in the efficacy of scuPA. PAI‐1‐TFT with DSP increases the efficacy of fibrinolytic therapy up to 8‐fold in chemically induced (sctPA and scuPA) and infectious (sctPA) pleural injury in rabbits. PAI‐1 is a valid molecular target in our model of S. pneumoniae empyema in rabbits, which closely recapitulates key characteristics of empyema in humans. Low‐dose PAI‐1‐TFT is a novel interventional strategy that offers the potential to improve fibrinolytic therapy for empyema in clinical practice., Empyema remains an important clinical problem with high mortality among patients 65 years and older. Targeting plasminogen activator inhibitor 1 (PAI‐1) in two rabbit models of pleural injury resulted in to up to eight‐fold increases in the efficacy of fibrinolytic therapy (decrease in dose needed for effective treatment). Low‐dose PAI‐1‐targeting fibrinolytic therapy would expand the population of patients that could benefit from treatment and improve therapeutic outcomes.
- Published
- 2021
34. Alpha‐defensins: risk factor for thrombosis in COVID‐19 infection
- Author
-
Suhair Abdeen, Douglas B. Cines, Charuhas Deshpande, Nigar Anjuman Khurram, Emad Maraga, Kathleen T. Montone, Leslie A. Litzky, Abd Al-Roof Higazi, Michael Feldman, Khalil Bdeir, Mohamed Higazi, Rami Abu-Fanne, and Samuel N. Heyman
- Subjects
Male ,Neutrophils ,medicine.medical_treatment ,Severity of Illness Index ,Mice ,0302 clinical medicine ,Risk Factors ,Prospective Studies ,biology ,Hematology ,Middle Aged ,Thrombosis ,Research Papers ,Tubulin Modulators ,Coagulation ,COVID‐19 infection ,030220 oncology & carcinogenesis ,Models, Animal ,Female ,medicine.symptom ,Research Paper ,Adult ,alpha-Defensins ,Antimicrobial peptides ,Inflammation ,Fibrin Fibrinogen Degradation Products ,03 medical and health sciences ,α‐defensins ,Thromboembolism ,Fibrinolysis ,medicine ,Animals ,Humans ,Risk factor ,Interleukin 6 ,Blood Coagulation ,thrombosis ,Aged ,business.industry ,Interleukin-6 ,SARS-CoV-2 ,COVID-19 ,medicine.disease ,interleukin‐6 ,Case-Control Studies ,Immunology ,biology.protein ,Neutrophil degranulation ,business ,Colchicine ,030215 immunology - Abstract
Summary The inflammatory response to SARS/CoV‐2 (COVID‐19) infection may contribute to the risk of thromboembolic complications. α‐Defensins, antimicrobial peptides released from activated neutrophils, are anti‐fibrinolytic and prothrombotic in vitro and in mouse models. In this prospective study of 176 patients with COVID‐19 infection, we found that plasma levels of α‐defensins were elevated, tracked with disease progression/mortality or resolution and with plasma levels of interleukin‐6 (IL‐6) and D‐dimers. Immunohistochemistry revealed intense deposition of α‐defensins in lung vasculature and thrombi. IL‐6 stimulated the release of α‐defensins from neutrophils, thereby accelerating coagulation and inhibiting fibrinolysis in human blood, imitating the coagulation pattern in COVID‐19 patients. The procoagulant effect of IL‐6 was inhibited by colchicine, which blocks neutrophil degranulation. These studies describe a link between inflammation and the risk of thromboembolism, and they identify a potential new approach to mitigate this risk in patients with COVID‐19 and potentially in other inflammatory prothrombotic conditions.
- Published
- 2021
35. SARS-CoV-2 Vaccine-Induced Immune Thrombotic Thrombocytopenia
- Author
-
James B. Bussel and Douglas B. Cines
- Subjects
2019-20 coronavirus outbreak ,COVID-19 Vaccines ,Coronavirus disease 2019 (COVID-19) ,business.industry ,SARS-CoV-2 ,viruses ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Vaccination ,virus diseases ,COVID-19 ,General Medicine ,030204 cardiovascular system & hematology ,Virology ,Thrombocytopenia ,03 medical and health sciences ,Editorial ,0302 clinical medicine ,Immune system ,ChAdOx1 nCoV-19 ,Pandemic ,Medicine ,Humans ,030212 general & internal medicine ,business - Abstract
The coronavirus disease 2019 (Covid-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) stimulated the development of highly effective vaccines that were produced wi...
- Published
- 2021
36. Thrombocytopenia following Pfizer and Moderna SARS-CoV-2 vaccination
- Author
-
Eun-Ju Lee, Craig M. Kessler, Bertrand Godeau, Marc Michel, Michele P. Lambert, Douglas B. Cines, Michael D. Tarantino, James B. Bussel, Terry Gernsheimer, John W. Semple, and Donald M. Arnold
- Subjects
Adult ,Male ,2019-20 coronavirus outbreak ,COVID-19 Vaccines ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Platelet Transfusion ,Young Adult ,Adrenal Cortex Hormones ,Commentaries ,Medicine ,Humans ,BNT162 Vaccine ,Aged ,Purpura, Thrombocytopenic, Idiopathic ,business.industry ,Platelet Count ,Incidence ,COVID-19 ,Immunoglobulins, Intravenous ,Hematology ,Middle Aged ,Virology ,Vaccination ,Platelet transfusion ,Commentary ,Female ,business ,2019-nCoV Vaccine mRNA-1273 - Published
- 2021
37. Mesenchymal stromal cells enhance self-assembly of a HUVEC tubular network through uPA-uPAR/VEGFR2/integrin/NOTCH crosstalk
- Author
-
I. B. Beloglazova, Natalia V Koptelova, Pyotr A. Tyurin-Kuzmin, Daniyar T. Dyikanov, Douglas B. Cines, Andrew P. Mazar, Victoria Stepanova, O. S. Plekhanova, Yelena V. Parfyonova, K. V. Dergilev, and E. S. Zubkova
- Subjects
Angiogenesis ,Integrin ,Notch signaling pathway ,Neovascularization, Physiologic ,Receptors, Urokinase Plasminogen Activator ,Extracellular matrix ,Human Umbilical Vein Endothelial Cells ,Matrix Metalloproteinase 14 ,Humans ,Receptor ,Molecular Biology ,Cells, Cultured ,biology ,Receptors, Notch ,Chemistry ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Cell Biology ,LRP1 ,Urokinase-Type Plasminogen Activator ,Vascular Endothelial Growth Factor Receptor-2 ,Cell biology ,Fibronectins ,Urokinase receptor ,CD18 Antigens ,cardiovascular system ,biology.protein ,Signal Transduction - Abstract
Endothelial cells (ECs) degrade the extracellular matrix of vessel walls and contact surrounding cells to facilitate migration during angiogenesis, leading to formation of an EC-tubular network (ETN). Mesenchymal stromal cells (MSC) support ETN formation when co-cultured with ECs, but the mechanism is incompletely understood. We examined the role of the urokinase-type plasminogen activator (uPA) system, i.e. the serine protease uPA, its inhibitor PAI-1, receptor uPAR/CD87, clearance by the low-density lipoprotein receptor-related protein (LRP1) and their molecular partners, in the formation of ETNs supported by adipose tissue-derived MSC. Co-culture of human umbilical vein ECs (HUVEC) with MSC increased mRNA expression levels of uPAR, MMP14, VEGFR2, TGFβ1, integrin β3 and Notch pathway components (Notch1 receptor and ligands: Dll1, Dll4, Jag1) in HUVECs and uPA, uPAR, TGFβ1, integrin β3, Jag1, Notch3 receptor in MSC. Inhibition at several steps in the activation process indicates that uPA, uPAR and LRP1 cross-talk with αv-integrins, VEGFR2 and Notch receptors/ligands to mediate ETN formation in HUVEC-MSC co-culture. The urokinase system mediates ETN formation through the coordinated action of uPAR, uPA's catalytic activity, its binding to uPAR and its nuclear translocation. These studies identify potential targets to help control aberrant angiogenesis with minimal impact on healthy vasculature.
- Published
- 2020
38. COVID-19 vaccination and immune thrombocytopenia
- Author
-
Douglas B. Cines, Allyson M. Pishko, and James B. Bussel
- Subjects
0301 basic medicine ,2019-20 coronavirus outbreak ,Coronavirus disease 2019 (COVID-19) ,business.industry ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,General Medicine ,Causality ,General Biochemistry, Genetics and Molecular Biology ,Immune thrombocytopenia ,Vaccination ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,030220 oncology & carcinogenesis ,Immunology ,Medicine ,Prospective cohort study ,business - Abstract
A prospective cohort analysis finds a link between the ChAdOx1 vaccine and an autoimmune disorder known as immune thrombocytopenia—but questions remain and causality is yet to be established.
- Published
- 2021
39. COVID-19-associated Acute Respiratory Distress Syndrome Clarified: A Vascular Endotype?
- Author
-
John P. Reilly, Douglas B. Cines, Nuala J. Meyer, Andrew E. Vaughan, Christopher A. Hunter, and Nilam S. Mangalmurti
- Subjects
Pulmonary and Respiratory Medicine ,2019-20 coronavirus outbreak ,Endotype ,Pulmonary Circulation ,Coronavirus disease 2019 (COVID-19) ,Pneumonia, Viral ,Acute respiratory distress ,Critical Care and Intensive Care Medicine ,Betacoronavirus ,Pandemic ,Correspondence ,Medicine ,Humans ,Pandemics ,Respiratory Distress Syndrome ,biology ,business.industry ,Viral Epidemiology ,SARS-CoV-2 ,COVID-19 ,medicine.disease ,biology.organism_classification ,Pneumonia ,Immunology ,business ,Coronavirus Infections - Published
- 2020
40. FcRn augments induction of tissue factor activity by IgG-containing immune complexes
- Author
-
Mortimer Poncz, Amrita Sarkar, Douglas B. Cines, Alan E. Mast, Sriram Krishnaswamy, Jonathan J. Hubbard, Keith R. McCrae, M. Anna Kowalska, Guohua Zhao, Richard S. Blumberg, Victoria Stepanova, Lubica Rauova, Laurence J. Blumberg, Michal Pyzik, Ann H. Rux, and Sergei Zaitsev
- Subjects
0301 basic medicine ,Male ,medicine.drug_class ,Immunology ,Antigen-Antibody Complex ,Receptors, Fc ,030204 cardiovascular system & hematology ,Monoclonal antibody ,Antibodies, Monoclonal, Humanized ,Platelet Factor 4 ,Biochemistry ,Immunoglobulin G ,Monocytes ,Thrombosis and Hemostasis ,Thromboplastin ,03 medical and health sciences ,Tissue factor ,Mice ,0302 clinical medicine ,Neonatal Fc receptor ,Immune system ,medicine ,Animals ,Humans ,biology ,Chemistry ,Heparin ,Histocompatibility Antigens Class I ,Anticoagulants ,Cell Biology ,Hematology ,Thrombocytopenia ,030104 developmental biology ,IgG binding ,biology.protein ,Antibody ,Platelet factor 4 - Abstract
Thromboembolism complicates disorders caused by immunoglobulin G (IgG)–containing immune complexes (ICs), but the underlying mechanisms are incompletely understood. Prior evidence indicates that induction of tissue factor (TF) on monocytes, a pivotal step in the initiation, localization, and propagation of coagulation by ICs, is mediated through Fcγ receptor IIa (FcγRIIa); however, the involvement of other receptors has not been investigated in detail. The neonatal Fc receptor (FcRn) that mediates IgG and albumin recycling also participates in cellular responses to IgG-containing ICs. Here we asked whether FcRn is also involved in the induction of TF-dependent factor Xa (FXa) activity by IgG-containing ICs by THP-1 monocytic cells and human monocytes. Induction of FXa activity by ICs containing IgG antibodies to platelet factor 4 (PF4) involved in heparin-induced thrombocytopenia (HIT), β-2-glycoprotein-1 implicated in antiphospholipid syndrome, or red blood cells coated with anti-(α)-Rh(D) antibodies that mediate hemolysis in vivo was inhibited by a humanized monoclonal antibody (mAb) that blocks IgG binding to human FcRn. IgG-containing ICs that bind to FcγR and FcRn induced FXa activity, whereas IgG-containing ICs with an Fc engineered to be unable to engage FcRn did not. Infusion of an α-FcRn mAb prevented fibrin deposition after microvascular injury in a murine model of HIT in which human FcγRIIa was expressed as a transgene. These data implicate FcRn in TF-dependent FXa activity induced by soluble and cell-associated IgG-containing ICs. Antibodies to FcRn, now in clinical trials in warm autoimmune hemolytic anemia to lower IgG antibodies and IgG containing ICs may also reduce the risk of venous thromboembolism.
- Published
- 2020
41. Erythrocytes Reveal Complement Activation in Patients with COVID-19
- Author
-
Ariel R. Weisman, Leticia Kuri-Cervantes, Douglas B. Cines, Caroline A. G. Ittner, Nilam S. Mangalmurti, Michael R. Betts, E. John Wherry, Lk Metthew Lam, Wen-Chao Song, John D. Lambris, Nuala J. Meyer, John P. Reilly, Sophie J Murphy, and M. Betina Pampena
- Subjects
Male ,Erythrocytes ,Autopsy ,Disease ,Article ,Virus ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Mediator ,Sepsis ,Complement C4b ,medicine ,Humans ,Complement Activation ,030304 developmental biology ,0303 health sciences ,SARS-CoV-2 ,business.industry ,COVID-19 ,Middle Aged ,Peptide Fragments ,3. Good health ,Complement (complexity) ,Complement system ,Shock (circulatory) ,Complement C3b ,Immunology ,Female ,medicine.symptom ,Respiratory Insufficiency ,business ,030215 immunology - Abstract
COVID-19, the disease caused by the SARS-CoV-2 virus, can progress to multi-organ failure characterized by respiratory insufficiency, arrhythmias, thromboembolic complications and shock. The mortality of patients hospitalized with COVID-19 is unacceptably high and new strategies are urgently needed to rapidly identify and treat patients at risk for organ failure. Clinical epidemiologic studies demonstrate that vulnerability to organ failure is greatest after viral clearance from the upper airway, which suggests that dysregulation of the host immune response is a critical mediator of clinical deterioration and death. Autopsy and pre-clinical evidence implicate aberrant complement activation in endothelial injury and organ failure. A potential therapeutic strategy warranting investigation is to inhibit complement, with case reports of successful treatment of COVID-19 with inhibitors of complement. However, this approach requires careful balance between the host protective and potential injurious effects of complement activation, and biomarkers to identify the optimal timing and candidates for therapy are lacking. Here we report the presence of complement activation products on circulating erythrocytes from hospitalized COVID-19 patients using flow cytometry. These findings suggest that novel erythrocyte-based diagnostics provide a method to identify patients with dysregulated complement activation.
- Published
- 2020
42. Opposing effects of HNP1 (α-defensin-1) on plasma cholesterol and atherogenesis
- Author
-
Samuel N. Heyman, Mohamed Higazi, Rami Abu-Fanne, Aseel Masarwy, Douglas B. Cines, Emad Maraga, Abd Al-Roof Higazi, Suhair Abdeen, and Khalil Bdeir
- Subjects
0301 basic medicine ,Apolipoprotein E ,Physiology ,Neutrophils ,Mice, Knockout, ApoE ,030204 cardiovascular system & hematology ,Biochemistry ,Vascular Medicine ,Fats ,White Blood Cells ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,Animal Cells ,Blood plasma ,Medicine and Health Sciences ,Aorta ,Multidisciplinary ,Chemistry ,Anticholesteremic Agents ,Lipids ,Body Fluids ,Lipoproteins, LDL ,Cholesterol ,Blood ,Medicine ,Female ,lipids (amino acids, peptides, and proteins) ,Anatomy ,Cellular Types ,medicine.symptom ,Research Article ,Genetically modified mouse ,alpha-Defensins ,medicine.medical_specialty ,Immune Cells ,Lipoproteins ,Science ,Immunology ,Cholestyramine Resin ,Mice, Transgenic ,Inflammation ,Diet, High-Fat ,Blood Plasma ,Lesion ,03 medical and health sciences ,Internal medicine ,medicine ,Animals ,Humans ,Nutrition ,Blood Cells ,Biology and Life Sciences ,Proteins ,Cell Biology ,Atherosclerosis ,Lipid Metabolism ,medicine.disease ,Diet ,Disease Models, Animal ,Hypocholesterolemia ,030104 developmental biology ,Endocrinology ,Colchicine ,Lipoprotein - Abstract
Atherosclerosis, the predominant cause of death in well-resourced countries, may develop in the presence of plasma lipid levels within the normal range. Inflammation may contribute to lesion development in these individuals, but the underlying mechanisms are not well understood. Transgenic mice expressing α-def-1 released from activated neutrophils develop larger lipid and macrophage-rich lesions in the proximal aortae notwithstanding hypocholesterolemia caused by accelerated clearance of α-def-1/low-density lipoprotein (LDL) complexes from the plasma. The phenotype does not develop when the release of α-def-1 is prevented with colchicine. However, ApoE-/- mice crossed with α-def-1 mice or given exogenous α-def-1 develop smaller aortic lesions associated with reduced plasma cholesterol, suggesting a protective effect of accelerated LDL clearance. Experiments were performed to address this seeming paradox and to determine if α-def-1 might provide a means to lower cholesterol and thereby attenuate atherogenesis. We confirmed that exposing ApoE-/- mice to α-def-1 lowers total plasma cholesterol and decreases lesion size. However, lesion size was larger than in mice with total plasma cholesterol lowered to the same extent by inhibiting its adsorption or by ingesting a low-fat diet. Furthermore, α-def-1 levels correlated independently with lesion size in ApoE-/- mice. These studies show that α-def-1 has competing effects on atherogenesis. Although α-def-1 accelerates LDL clearance from plasma, it also stimulates deposition and retention of LDL in the vasculature, which may contribute to development of atherosclerosis in individuals with normal or even low plasma levels of cholesterol. Inhibiting α-def-1 may attenuate the impact of chronic inflammation on atherosclerotic vascular disease.
- Published
- 2020
43. American Society of Hematology 2018 guidelines for management of venous thromboembolism: heparin-induced thrombocytopenia
- Author
-
Reem A. Mustafa, Andreas Greinacher, Theodore E. Warkentin, Stephen B. Rodner, Sixten Selleng, Rebecca L. Morgan, Beng H. Chong, Yves Gruel, Lori A. Linkins, Gowthami M. Arepally, Adam Cuker, Ashleigh Wex, Douglas B. Cines, and Nancy Santesso
- Subjects
medicine.medical_specialty ,Danaparoid ,Administration, Oral ,Dermatan Sulfate ,030204 cardiovascular system & hematology ,Arginine ,Fondaparinux ,Argatroban ,03 medical and health sciences ,0302 clinical medicine ,Heparin-induced thrombocytopenia ,medicine ,Humans ,Bivalirudin ,030212 general & internal medicine ,Intensive care medicine ,Sulfonamides ,Evidence-Based Medicine ,Heparin ,Platelet Count ,business.industry ,Cardiovascular Surgical Procedures ,Chondroitin Sulfates ,Anticoagulants ,Venous Thromboembolism ,Hematology ,Evidence-based medicine ,Guideline ,Hirudins ,medicine.disease ,Thrombocytopenia ,Peptide Fragments ,Recombinant Proteins ,Renal Replacement Therapy ,Pre- and post-test probability ,Pipecolic Acids ,Heparitin Sulfate ,business ,Clinical Guidelines ,medicine.drug - Abstract
Background: Heparin-induced thrombocytopenia (HIT) is an adverse drug reaction mediated by platelet-activating antibodies that target complexes of platelet factor 4 and heparin. Patients are at markedly increased risk of thromboembolism. Objective: These evidence-based guidelines of the American Society of Hematology (ASH) are intended to support patients, clinicians, and other health care professionals in their decisions about diagnosis and management of HIT. Methods: ASH formed a multidisciplinary guideline panel balanced to minimize potential bias from conflicts of interest. The McMaster University GRADE Centre supported the guideline development process, including updating or performing systematic evidence reviews. The panel prioritized clinical questions and outcomes according to their importance for clinicians and patients. The Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach was used to assess evidence and make recommendations, which were subject to public comment. Results: The panel agreed on 33 recommendations. The recommendations address screening of asymptomatic patients for HIT, diagnosis and initial management of patients with suspected HIT, treatment of acute HIT, and special situations in patients with acute HIT or a history of HIT, including cardiovascular surgery, percutaneous cardiovascular intervention, renal replacement therapy, and venous thromboembolism prophylaxis. Conclusions: Strong recommendations include use of the 4Ts score rather than a gestalt approach for estimating the pretest probability of HIT and avoidance of HIT laboratory testing and empiric treatment of HIT in patients with a low-probability 4Ts score. Conditional recommendations include the choice among non-heparin anticoagulants (argatroban, bivalirudin, danaparoid, fondaparinux, direct oral anticoagulants) for treatment of acute HIT.
- Published
- 2018
44. T2 Magnetic Resonance to Monitor Hemostasis
- Author
-
Adam Cuker, Douglas B. Cines, Maria E. Carinato, Thomas Jay Lowery, and Joseph E. Marturano
- Subjects
Magnetic Resonance Spectroscopy ,Platelet Function Tests ,medicine.medical_treatment ,030204 cardiovascular system & hematology ,Fibrin ,03 medical and health sciences ,0302 clinical medicine ,Fibrinolysis ,Antithrombotic ,medicine ,Humans ,Platelet ,Whole blood ,Hemostasis ,medicine.diagnostic_test ,biology ,business.industry ,Magnetic resonance imaging ,Hematology ,medicine.disease ,Thrombosis ,biology.protein ,Cardiology and Cardiovascular Medicine ,business ,030215 immunology ,Biomedical engineering - Abstract
There is a clinical need for pragmatic approaches to measure integrated hemostatic reactions in whole blood rapidly, using small volumes of blood. The authors have applied T2 magnetic resonance (T2MR) to assess coagulation reactions based on partitioning of red blood cells and proteins that occurs during fibrin formation and platelet-mediated clot contraction. T2MR is amenable to measuring clotting times, individual coagulation factors, and platelet function. T2MR also revealed a novel “hypercoagulable” signature characterized by fibrin clots almost insusceptible to fibrinolysis that surround tessellated arrays of polyhedral erythrocytes (“third peak”). This signature, which develops under conditions associated with intense clot formation in vitro, may help identify patients at risk of developing thrombosis and for monitoring antithrombotic therapies in the future.
- Published
- 2018
45. Dynamic intercellular redistribution of HIT antigen modulates heparin-induced thrombocytopenia
- Author
-
Mortimer Poncz, Daria Madeeva, Valerie Tutwiler, Jing Dai, Douglas B. Cines, Vincent Hayes, Hyun Sook Ahn, Gowthami M. Arepally, and Lubica Rauova
- Subjects
Blood Platelets ,0301 basic medicine ,Immunology ,Monocytopenia ,030204 cardiovascular system & hematology ,Platelet Factor 4 ,Biochemistry ,Monocytes ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Heparin-induced thrombocytopenia ,Human Umbilical Vein Endothelial Cells ,medicine ,Animals ,Humans ,Platelet ,Antigens ,Whole blood ,Heparin ,Chemistry ,Monocyte ,Cell Biology ,Hematology ,medicine.disease ,Thrombocytopenia ,Molecular biology ,Endothelial stem cell ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Platelet factor 4 ,medicine.drug - Abstract
Heparin-induced thrombocytopenia (HIT) is a prothrombotic disorder initiated by antibodies to platelet factor 4 (PF4)/heparin complexes. PF4 released from platelets binds to surface glycosaminoglycans on hematopoietic and vascular cells that are heterogenous in composition and differ in affinity for PF4. PF4 binds to monocytes with higher affinity than to platelets, and depletion of monocytes exacerbates thrombocytopenia in a murine HIT model. Here we show that the expression of PF4 on platelets and development of thrombocytopenia are modulated by the (re)distribution of PF4 among hematopoietic and endothelial cell surfaces. Binding of PF4 to platelets in whole blood in vitro varies inversely with the white cell count, likely because of the greater affinity of monocytes for PF4. In mice, monocyte depletion increased binding of PF4 to platelets by two- to three-fold. Induction of HIT in mice caused a transient >80-fold increase in binding of HIT antibody to monocytes vs 3.5-fold increase to platelets and rapid transient monocytopenia. Normalization of monocyte counts preceded the return in platelet counts. Exposure of blood to endothelial cells also depletes PF4 from platelet surfaces. These studies demonstrate a dynamic interchange of surface-bound PF4 among hematopoetic and vascular cells that may limit thrombocytopenia at the expense of promoting prothrombotic processes in HIT.
- Published
- 2018
46. Management of antithrombotic therapy in adults with immune thrombocytopenia (ITP): a survey of ITP specialists and general hematologist–oncologists
- Author
-
Deirdra R. Terrell, Adam Cuker, Allyson M. Pishko, Mudi Misgav, James N. George, Sara K. Vesely, and Douglas B. Cines
- Subjects
Adult ,medicine.medical_specialty ,Medication Therapy Management ,medicine.drug_class ,030204 cardiovascular system & hematology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Fibrinolytic Agents ,immune system diseases ,Surveys and Questionnaires ,hemic and lymphatic diseases ,Internal medicine ,Antithrombotic ,medicine ,Humans ,In patient ,Practice Patterns, Physicians' ,Hematologist ,Oncologists ,Purpura, Thrombocytopenic, Idiopathic ,Hematology ,Platelet Count ,business.industry ,Anticoagulant ,Anticoagulants ,Oklahoma ,Middle Aged ,medicine.disease ,Thrombosis ,Optimal management ,Immune thrombocytopenia ,Cardiology and Cardiovascular Medicine ,business ,Platelet Aggregation Inhibitors ,Specialization ,030215 immunology - Abstract
While patients with immune thrombocytopenia (ITP) and low platelet counts are at risk for bleeding, they are not protected against arterial and venous thrombotic events. Frequently, hematologists are asked to consult on a patient with ITP requiring an antiplatelet (AP) agent or anticoagulant (AC). No direct evidence exists to guide hematologists in weighing the risk of thrombosis against the risk of bleeding in patients with ITP. Therefore, we performed a survey to determine the preferred management of AP/AC therapy in ITP patients. The survey described hypothetical patient scenarios and asked respondents to recommend a minimum platelet count for initiation of AP/AC therapy. We surveyed both hematologists with an international reputation in treatment of ITP (n = 48) and also general hematologist–oncologists in Oklahoma (n = 97). Response rates were 38/48 (79%) for the ITP specialists and 46/97 (47%) for general hematologist–oncologists. Overall, recommended platelet thresholds for antithrombotic therapy were similar between ITP specialists and general hematologist–oncologists. Although both groups recommended a minimum platelet count of 50 × 109/L for AP and AC therapy in most scenarios, there was great variability in individual practice patterns among respondents. This study highlights the need for studies of patients with ITP who require AP/AC therapy to provide high-quality evidence for establishing optimal management strategies.
- Published
- 2018
47. Release of IL-6 After Stroke Contributes to Impaired Cerebral Autoregulation and Hippocampal Neuronal Necrosis Through NMDA Receptor Activation and Upregulation of ET-1 and JNK
- Author
-
Hugh Hekierski, Abd Al-Roof Higazi, Philip Pastor, Serge Yarovoi, Douglas B. Cines, and William M. Armstead
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Swine ,medicine.medical_treatment ,Inflammation ,Vasodilation ,Hippocampus ,Receptors, N-Methyl-D-Aspartate ,Cerebral autoregulation ,Necrosis ,Random Allocation ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Internal medicine ,Fibrinolysis ,Animals ,Homeostasis ,Medicine ,cardiovascular diseases ,Stroke ,Oxazolidinones ,Anthracenes ,Cerebral Cortex ,Endothelin-1 ,Interleukin-6 ,business.industry ,General Neuroscience ,medicine.disease ,Endothelin 1 ,Up-Regulation ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,Tissue Plasminogen Activator ,Neurology (clinical) ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business ,030217 neurology & neurosurgery ,Vasoconstriction ,Signal Transduction - Abstract
The sole FDA-approved drug treatment for ischemic stroke is tissue-type plasminogen activator (tPA). However, upregulation of JNK mitogen-activated protein kinase (MAPK) and endothelin 1 (ET-1) by tPA after stroke contributes to impaired cerebrovascular autoregulation. Wild-type (wt) tPA can bind to the lipoprotein-related receptor (LRP), which mediates vasodilation, or NMDA receptors (NMDA-Rs), exacerbating vasoconstriction. Elevations in IL-6, a marker of inflammation that accompanies stroke, are reported to be an adverse prognostic factor. We hypothesized that IL-6 released into CSF after stroke by wt-tPA through activation of NMDA-Rs and upregulation of ET-1 and JNK contribute to impairment of cerebrovascular autoregulation and increased histopathology. Results show that IL-6 was increased post stroke in pigs, which was increased further by wt-tPA. Co-administration of the IL-6 antagonist LMT-28 with wt-tPA prevented impairment of cerebrovascular autoregulation and necrosis of hippocampal cells. wt-tPA co-administered with the JNK inhibitor SP 600125 and the ET-1 antagonist BQ 123 blocked stroke-induced elevation of IL-6. Co-administration of LMT-28 with wt-tPA blocked the augmentation of JNK and ET-1 post stroke. In conclusion, IL-6 released after stroke, which is enhanced by wt-tPA through activation of NMDA-Rs and upregulation of ET-1 and JNK, impairs cerebrovascular autoregulation and increases histopathology. Strategies that promote fibrinolysis while limiting activation of NMDA-Rs and upregulation of IL-6 may improve the benefit/risk ratio compared to wt-tPA in treatment of stroke.
- Published
- 2018
48. Biocompatible coupling of therapeutic fusion proteins to human erythrocytes
- Author
-
Elizabeth D. Hood, Mortimer Poncz, Carlos H. Villa, Ian Johnston, Vladimir R. Muzykantov, Douglas B. Cines, Daniel C. Pan, Colin F. Greineder, Landis R. Walsh, and Don L. Siegel
- Subjects
0301 basic medicine ,Erythrocytes ,Recombinant Fusion Proteins ,Thrombomodulin ,chemical and pharmacologic phenomena ,Epitope ,Mice ,03 medical and health sciences ,Drug Delivery Systems ,Red Cells, Iron, and Erythropoiesis ,0302 clinical medicine ,Thrombin ,stomatognathic system ,Anion Exchange Protein 1, Erythrocyte ,medicine ,Animals ,Humans ,Glycophorin ,Band 3 ,Inflammation ,Rh-Hr Blood-Group System ,biology ,Chemistry ,Immunogenicity ,Thrombosis ,Hematology ,respiratory system ,medicine.disease ,Fusion protein ,Molecular biology ,Hemolysis ,030104 developmental biology ,030220 oncology & carcinogenesis ,cardiovascular system ,biology.protein ,Macaca ,sense organs ,Single-Chain Antibodies ,medicine.drug - Abstract
Carriage of drugs by red blood cells (RBCs) modulates pharmacokinetics, pharmacodynamics, and immunogenicity. However, optimal targets for attaching therapeutics to human RBCs and adverse effects have not been studied. We engineered nonhuman-primate single-chain antibody fragments (scFvs) directed to human RBCs and fused scFvs with human thrombomodulin (hTM) as a representative biotherapeutic cargo (hTM-scFv). Binding fusions to RBCs on band 3/glycophorin A (GPA; Wright b [Wrb] epitope) and RhCE (Rh17/Hr0 epitope) similarly endowed RBCs with hTM activity, but differed in their effects on RBC physiology. scFv and hTM-scFv targeted to band 3/GPA increased membrane rigidity and sensitized RBCs to hemolysis induced by mechanical stress, while reducing sensitivity to hypo-osmotic hemolysis. Similar properties were seen for other ligands bound to GPA and band 3 on human and murine RBCs. In contrast, binding of scFv or hTM-scFv to RhCE did not alter deformability or sensitivity to mechanical and osmotic stress at similar copy numbers bound per RBCs. Contrasting responses were also seen for immunoglobulin G antibodies against band 3, GPA, and RhCE. RBC-bound hTM-scFv generated activated protein C (APC) in the presence of thrombin, but RhCE-targeted hTM-scFv demonstrated greater APC generation per bound copy. Both Wrb- and RhCE-targeted fusion proteins inhibited fibrin deposition induced by tumor necrosis factor-α in an endothelialized microfluidic model using human whole blood. RhCE-bound hTM-scFv more effectively reduced platelet and leukocyte adhesion, whereas anti-Wrb scFv appeared to promote platelet adhesion. These data provide a translational framework for the development of engineered affinity ligands to safely couple therapeutics to human RBCs.
- Published
- 2018
49. Targeting plasminogen activator inhibitor-1 in tetracycline-induced pleural injury in rabbits
- Author
-
Ali Azghani, Steven Idell, Chris Schaefer, Douglas B. Cines, Serge Yarovoi, Paul Declerck, Sophia Karandashova, Andrey A. Komissarov, and Galina Florova
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Pathology ,medicine.medical_specialty ,Physiology ,Tetracycline ,medicine.medical_treatment ,Pharmacology ,03 medical and health sciences ,chemistry.chemical_compound ,Fibrinolytic Agents ,Physiology (medical) ,Plasminogen Activator Inhibitor 1 ,Fibrinolysis ,Animals ,Medicine ,Thrombolytic Therapy ,Respiratory system ,Adverse effect ,Protein Synthesis Inhibitors ,Protein synthesis inhibitor ,business.industry ,Cell Biology ,Pleural Diseases ,Disease Models, Animal ,030104 developmental biology ,chemistry ,Plasminogen activator inhibitor-1 ,Female ,Rabbits ,business ,Plasminogen activator ,Fibrinolytic agent ,Research Article ,medicine.drug - Abstract
Elevated active plasminogen activator inhibitor-1 (PAI-1) has an adverse effect on the outcomes of intrapleural fibrinolytic therapy (IPFT) in tetracycline-induced pleural injury in rabbits. To enhance IPFT with prourokinase (scuPA), two mechanistically distinct approaches to targeting PAI-1 were tested: slowing its reaction with urokinase (uPA) and monoclonal antibody (mAb)-mediated PAI-1 inactivation. Removing positively charged residues at the “PAI-1 docking site” (179RHRGGS184→179AAAAAA184) of uPA results in a 60-fold decrease in the rate of inhibition by PAI-1. Mutant prourokinase (0.0625–0.5 mg/kg; n = 12) showed efficacy comparable to wild-type scuPA and did not change IPFT outcomes ( P > 0.05). Notably, the rate of PAI-1-independent intrapleural inactivation of mutant uPA was 2 times higher ( P < 0.05) than that of the wild-type enzyme. Trapping PAI-1 in a “molecular sandwich”-type complex with catalytically inactive two-chain urokinase with Ser195Ala substitution (S195A-tcuPA; 0.1 and 0.5 mg/kg) did not improve the efficacy of IPFT with scuPA (0.0625–0.5 mg/kg; n = 11). IPFT failed in the presence of MA-56A7C10 (0.5 mg/kg; n = 2), which forms a stable intrapleural molecular sandwich complex, allowing active PAI-1 to accumulate by blocking its transition to a latent form. In contrast, inactivation of PAI-1 by accelerating the active-to-latent transition mediated by mAb MA-33B8 (0.5 mg/kg; n = 2) improved the efficacy of IPFT with scuPA (0.25 mg/kg). Thus, under conditions of slow (4–8 h) fibrinolysis in tetracycline-induced pleural injury in rabbits, only the inactivation of PAI-1, but not a decrease in the rate of its reaction with uPA, enhances IPFT. Therefore the rate of fibrinolysis, which varies in different pathologic states, could affect the selection of PAI-1 inhibitors to enhance fibrinolytic therapy.
- Published
- 2018
50. Sars-Cov-2 Vaccination in Patients with Pre-Existing Immune Thrombocytopenia
- Author
-
Eun-Ju Lee, Douglas B. Cines, Howard A. Liebman, Jecko Thachil, Craig M. Kessler, Alexandra Kruse, Michael D. Tarantino, Ashley Ray, Adrian C. Newland, Adam Cuker, Marina Beltrami Moreira, Andrew D. Leavitt, Alfred Ian Lee, Hanny Al-Samkari, Terry B. Gernsheimer, Caroline Kruse, Jennifer DiRaimo, David J. Kuter, and James B. Bussel
- Subjects
311.Disorders of Platelet Number or Function: Clinical and Epidemiological ,business.industry ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Immunology ,education ,Cell Biology ,Hematology ,Biochemistry ,Immune thrombocytopenia ,Vaccination ,Medicine ,In patient ,business ,health care economics and organizations - Abstract
Introduction: Cases of de novo immune thrombocytopenia (ITP), including a fatality following SARS-CoV-2 vaccination in a previously healthy recipient, led to studying its impact in pre-existing ITP. Published reports are limited but suggest that most patients with ITP tolerate the COVID-19 vaccines well without frequent ITP exacerbations (Kuter, BJH, 2021). Data regarding risk factors for exacerbation and relationship of response to first dose to that of second dose are limited. Methods: Data for patients with pre-existing ITP were obtained via 3 sources. First, via a ten-center retrospective study of adults with ITP who received a SARS-CoV-2 vaccine between December 2020 and March 2021 and had a post-vaccination platelet count (n=117); 9 centers were in the United States. Eighty-nine percent of patients received mRNA-based vaccines. The second and third sources of data were surveys distributed by the Platelet Disorder Support Association (PDSA) and the United Kingdom ITP Support Association. A 'stable platelet count' was defined as a post-vaccination platelet count within 20% of the pre-vaccination level. ITP exacerbation was defined as any one or more of: platelet decrease ≥ 50% compared to pre-vaccination baseline, platelet decrease by >20% compared to pre-vaccination baseline with platelet nadir < 30x10 9/L, receipt of rescue therapy for ITP. Continuous variables were described as mean ±SD or median [interquartile range]; categorical variables were described as n (%). Relative risks and 95% confidence interval were calculated to estimate strength of association. Results: Among 117 patients with pre-existing ITP from 10 centers who received a SARS-CoV-2 vaccine, mean age was 63±17 years, 62% were female, with median 12 [4-23] years since diagnosis of ITP; patients had received a median of 3 [2-4] prior medical treatments. Sixty-nine patients were on ITP treatment at the time of vaccination (Table 1). There was an almost even distribution of platelet count response following each vaccine dose. In 109 patients with data for dose 1, platelet counts increased in 32 (29%), were stable in 43 (39%), and decreased in 34 (31%); in 70 patients following dose 2, platelet counts increased in 24 (34%), were stable in 25 (36%), and decreased in 21 (30%) (Figure 1). Nineteen (17%) patients experienced an ITP exacerbation following the first dose and 14 (20%) of 70 after a second dose. In total, fifteen patients received and responded to rescue treatments (n = 6 after dose 1, n = 8 after dose 2, n = 1 after both doses). Of 7 patients who received rescue treatment after dose 1, 5 received dose 2 and only 1/5 received rescue treatment again. Rescue consisted of increased dose of ongoing medication, steroids, IVIG, and rituximab. Splenectomized persons and those who received 5 or more prior lines of medical therapy were at highest risk of ITP exacerbation. Only 1 of 47 patients who had neither undergone splenectomy nor received 5 or more lines of therapy developed ITP exacerbation after dose 1. There were 14 patients off-treatment at the time of dose 1 and 7 patients at time of dose 2; 1 patient in each group developed ITP exacerbation with both these having had normal counts prior to vaccination and having undergone splenectomy. In 43 patients whose platelet counts were stable or increased after dose 1 and received dose 2, only 6 (14%) had platelet decreases to Conclusions: Thrombocytopenia may worsen in pre-existing ITP post-SARS-CoV2-vaccination but when ITP exacerbation occurred, it responded well to rescue treatment. No serious bleeding events were noted. Rescue treatment was needed in 13% of patients. Proactive vaccination surveillance of patients with known ITP, especially those post-splenectomy and with more refractory disease, is indicated. These findings should encourage patients with ITP to not only be vaccinated, but to receive the second dose when applicable to ensure optimal immunization. Rituximab interferes with vaccination response and ideally would be held until a minimum of 2 weeks after completion of vaccination. Figure 1 Figure 1. Disclosures Lee: Principia Biopharma: Consultancy. Beltrami Moreira: Kadmon: Other: Spouse current employer; Jounce Therapeutics: Other: Spouse employment ended in the past 24 months. Al-Samkari: Amgen: Research Funding; Argenx: Consultancy; Agios: Consultancy, Research Funding; Dova: Consultancy, Research Funding; Rigel: Consultancy; Sobi: Consultancy; Novartis: Consultancy. Cuker: Novo Nordisk: Research Funding; Pfizer: Research Funding; Bayer: Research Funding; Sanofi: Research Funding; Takeda: Research Funding; Novartis: Research Funding; UpToDate: Patents & Royalties; Alexion: Research Funding; Spark Therapeutics: Research Funding; Synergy: Consultancy. MacWhirter - DiRaimo: JD has not personally received any payment personally, but PDSA has received grants and consultancy fees from Novartis, grant and honorarium from Amgen, grant and consultancy fees from Pfizer and UCB, and grants from Argenx, Principia, Rigel, and CSL Behr: Consultancy, Honoraria, Other. Gernsheimer: Cellphire: Consultancy; Rigel: Research Funding; Principia: Research Funding; Novartis: Honoraria; Amgen: Honoraria; Dova: Consultancy; Sanofi: Consultancy. Kruse: Alex has not personally received any payment but reports that PDSA received grants and consultancy fees from Novartis, grant and honorarium from Amgen, grant and consultancy fees from Pfizer and UCB, and grants from Argenx, Principia, Rigel, and CSL Behri: Consultancy, Honoraria, Other. Kessler: Genentech: Membership on an entity's Board of Directors or advisory committees, Research Funding; Octapharma: Membership on an entity's Board of Directors or advisory committees, Research Funding; Bayer: Membership on an entity's Board of Directors or advisory committees, Research Funding; Takeda: Membership on an entity's Board of Directors or advisory committees, Research Funding; Novo Nordisk: Membership on an entity's Board of Directors or advisory committees; Pfizer: Membership on an entity's Board of Directors or advisory committees; CSL Behring: Membership on an entity's Board of Directors or advisory committees. Kruse: CK has not personally received payment but reports that PDSA received grants and consultancy fees from Novartis, grant and honorarium from Amgen, grant and consultancy fees from Pfizer and UCB, and grants from Argenx, Principia, Rigel, and CSL Behring: Consultancy, Honoraria, Other. Leavitt: Pfizer: Research Funding; Merck: Consultancy; CSL DOVA: Consultancy; Catalys: Consultancy; Behring: Consultancy; BPL: Consultancy; HEMA Biologics: Consultancy; Rigel: Consultancy; Syntimmune: Research Funding; Sangamo Therapeutics: Research Funding; BioMarin: Consultancy, Research Funding. Liebman: Sanofi/Genzyme: Research Funding; Novartis: Consultancy, Research Funding; Argenx: Research Funding; Amgen: Consultancy; Dova: Consultancy, Honoraria; Pfizer: Consultancy. Newland: Novartis: Consultancy, Research Funding, Speakers Bureau; UCB Biosciences: Consultancy; Roche: Speakers Bureau; Octapharma: Research Funding; GSK: Consultancy, Research Funding, Speakers Bureau; BMS: Research Funding; Amgen: Consultancy, Research Funding, Speakers Bureau; Argenx: Consultancy, Speakers Bureau; Angle: Consultancy; Grifols: Consultancy, Speakers Bureau. Tarantino: Pfizer: Research Funding; Novo Nordisk: Research Funding; Takeda: Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; UCB: Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Grifols: Research Funding, Speakers Bureau; Principia: Membership on an entity's Board of Directors or advisory committees, Research Funding; Spark Therapeutics: Membership on an entity's Board of Directors or advisory committees, Research Funding; Sobi: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Amgen: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Dova: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Octapharma: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; BioMarin: Membership on an entity's Board of Directors or advisory committees; Genentech: Membership on an entity's Board of Directors or advisory committees. Thachil: Amgen: Speakers Bureau; Novartis: Speakers Bureau. Kuter: Up-to-Date: Patents & Royalties: Up-To-Date; Platelet Disorder Support Association: Membership on an entity's Board of Directors or advisory committees; Actelion (Syntimmune), Agios, Alnylam, Amgen, Argenx, BioCryst, Bristol Myers Squibb (BMS), Caremark, CRICO, Daiichi Sankyo, Dova, Genzyme, Immunovant, Incyte, Kyowa-Kirin, Merck Sharp Dohme, Momenta, Novartis, Pfizer, Principia, Protalex, Protalix, Rigel: Consultancy, Other: grant support and consulting fees; Actelion (Syntimmune), Agios, Alnylam, Amgen, Argenx, Bristol Myers Squibb (BMS), Immunovant, Kezar, Principia, Protalex, Rigel, Takeda (Bioverativ), UCB: Research Funding; Rubius: Current equity holder in publicly-traded company. Cines: Dova: Consultancy; Rigel: Consultancy; Treeline: Consultancy; Arch Oncol: Consultancy; Jannsen: Consultancy; Taventa: Consultancy; Principia: Other: Data Safety Monitoring Board. Bussel: CSL: Other: DSMB; Dova/Sobi: Consultancy, Membership on an entity's Board of Directors or advisory committees; UCB: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: DSMB; Momenta/Janssen: Consultancy, Membership on an entity's Board of Directors or advisory committees; Amgen: Consultancy, Membership on an entity's Board of Directors or advisory committees; Argenx: Consultancy, Membership on an entity's Board of Directors or advisory committees; UptoDate: Honoraria; RallyBio: Consultancy, Membership on an entity's Board of Directors or advisory committees; Principia/Sanofi: Consultancy, Membership on an entity's Board of Directors or advisory committees; Rigel: Consultancy, Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees.
- Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.