Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Clin Appl Thromb Hemost ; 29: 10760296231166370, 2023.
Article in English | MEDLINE | ID: mdl-37069794

ABSTRACT

BACKGROUND: Heparin-induced thrombocytopenia (HIT) is a common differential diagnosis in cardiothoracic surgery. The latex immunoturbidimetric assay (LIA) is an enhanced immunoassay that has recently been introduced for the detection of total HIT immunoglobulin and retains a higher specificity of 95% compared to the enzyme-linked immunosorbent assay. OBJECTIVES: To investigate if a semiquantitative relationship exists between increasing LIA levels beyond the current positivity threshold and its correlation to positive serotonin release assay results in cardiothoracic surgery. METHODS: This was a multicenter, observational cohort of cardiothoracic surgery patients initiated on anticoagulation with heparin-based products. To conduct sensitivity and specificity analysis of LIA values, HIT positive was defined as a LIA value ≥1 unit/mL and HIT negative was defined as a LIA level <1 unit/mL. A receiver operating characteristic (ROC) analysis was utilized to evaluate the predictive performance of the LIA. RESULTS: At manufactures' cutoffs of ≥1.0 unit/mL, LIA sensitivity and specificity was 93.8% and 22%, respectively, yielding a false positive rate of 78%. At a higher cutoff of 4.5 units/mL, LIA sensitivity and specificity was 75% and 71%, respectively, yielding a false positive rate of 29% and an area under the ROC curve of 0.75 (P = .01; 95% confidence interval: 0.621-0.889). Bivalirudin was initiated in 84.6% of false positive LIA results. CONCLUSION: This study suggests that the diagnostic accuracy of the LIA can be optimized by increasing the LIA positivity threshold. Proposing a higher LIA cutoff, may mitigate unwarranted anticoagulation and bleeding outcomes.


Subject(s)
Latex , Thrombocytopenia , Humans , Latex/adverse effects , Immunoturbidimetry , Thrombocytopenia/chemically induced , Thrombocytopenia/diagnosis , Heparin/adverse effects , Enzyme-Linked Immunosorbent Assay/methods , Anticoagulants/adverse effects
2.
Int J Lab Hematol ; 41(4): 478-484, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30986338

ABSTRACT

INTRODUCTION: Heparin-induced thrombocytopenia (HIT) is a potentially life-threatening adverse reaction of heparin. Laboratory evaluation of HIT is often not available within a reasonable time. We evaluated the HemosIL® HIT-Ab(PF4-H) (Instrumentation Laboratory), a rapid, on-demand, fully automated, latex immunoturbidimetric assay (LIA). MATERIALS AND METHODS: Following determination of the LIA's reference interval and cutoff values, a multicenter study was conducted between March 2013 and June 2015. Plasma samples of HIT-suspected patients (n = 632) were collected and evaluated by LIA on the ACL TOP® Family systems (Instrumentation Laboratory), enzyme-linked immunosorbent assays (EIA), and serotonin release assay (SRA). Patient characteristics, medical conditions, comorbidities, laboratory results, and medications were collected via medical chart review. The pretest clinical probability of HIT was also calculated for each patient. RESULTS: Based on the 95% reference interval for healthy donors and HIT-negative patients, a LIA value ≥1.0 U/mL was interpreted positive. The overall agreement of LIA versus EIA and SRA results were 90% (95% CI 88%-92%) and 79% (95% CI 75%-82%), respectively. The negative predictive value for LIA and EIA was comparable (87%) with SRA. The positive and negative percent agreements with the clinical probability were 89% (95% CI 69%-97%) and 86% (95% CI 83%-89%), respectively, with a negative predictive value of 99.6% (95% CI 98%-100%). DISCUSSION: Overall, the LIA results were comparable to those of EIA and SRA. This fully automated assay with a remarkable short analytical turnaround time of <20 minutes can be performed on-demand, which would greatly facilitate more prompt management of HIT.


Subject(s)
Heparin/adverse effects , Serotonin/blood , Thrombocytopenia , Enzyme-Linked Immunosorbent Assay , Female , Heparin/administration & dosage , Humans , Male , Middle Aged , Thrombocytopenia/blood , Thrombocytopenia/chemically induced , Thrombocytopenia/diagnosis
3.
Article in English | MEDLINE | ID: mdl-27956417

ABSTRACT

Previous studies have shown that some lipoglycopeptide and lipopeptide antimicrobial agents may cause falsely elevated values for some phospholipid-dependent coagulation tests. The effect of oritavancin, a lipoglycopeptide antibiotic, on coagulation test results was explored using pooled human plasma samples spiked with drug and in a clinical study after an infusion of a single 1,200-mg intravenous dose of oritavancin in normal healthy volunteers. Pooled plasma with oritavancin added ex vivo showed concentration-dependent prolongation of prothrombin time/international normalized ratio (PT/INR), activated partial thromboplastin time (aPTT), and dilute Russell viper venom time (DRVVT) test results. In contrast, oritavancin had no effect on the activated protein C resistance assay, chromogenic anti-factor Xa assay (anti-FXa), thrombin time, and an immunoassay for the laboratory diagnosis of heparin-induced thrombocytopenia. In participants that received a single dose of oritavancin, elevations in PT/INR result, aPTT, DRVVT, activated clotting time, and silica clotting time occurred, with the maximum times to resolution of test interference determined to be 12, 120, 72, 24, and 18 h, respectively. The anti-FXa assay was unaffected, whereas transient elevations in D dimer levels were observed in 30% of participants, with a maximum time to resolution of 72 h. Although oritavancin has no impact on the coagulation system in vivo, a single dose of oritavancin can produce falsely elevated values of some coagulation tests used to monitor hemostasis. The interference of oritavancin on affected tests is transient, and the test results revert to normal ranges within specified times after dosing.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Blood Coagulation/drug effects , Glycopeptides/therapeutic use , Adult , Blood Coagulation Tests , Female , Hemostasis/drug effects , Humans , Lipoglycopeptides , Male , Middle Aged , Young Adult
5.
Thromb Res ; 141: 39-48, 2016 05.
Article in English | MEDLINE | ID: mdl-26967531

ABSTRACT

BACKGROUND: Tissue factor (TF) is involved in tumor growth and metastasis and contributes to venous thromboembolism (VTE) in cancer, including gynecological malignancies. The diagnostic value of microvesicle-associated TF procoagulant activity (MV TF PCA) in women with suspected ovarian cancer, however, has not been studied. OBJECTIVE: To evaluate MV TF PCA as a diagnostic tool in women with an ovarian mass of unknown etiology and as a predictive biomarker for perioperative VTE. METHODS: Plasma MVs were isolated by high-speed centrifugation and analyzed for TF-specific PCA by single-stage clotting assay. In addition, plasma TF antigen and soluble P-selectin (sCD62P) were measured by ELISA. RESULTS: D-Dimer, MV TF PCA, and sCD62P, but not the tumor marker, CA-125, significantly differentiated patients with malignant (n=40) from those with benign tumors (n=15) and healthy controls (n=34). In cancer patients, only D-Dimer and CA-125 correlated with the FIGO stage. An abnormal D-dimer had the highest sensitivity for the diagnosis of cancer, while MV TF PCA above the ROC curve-derived cut-off value of 182U/mL had the highest specificity. By multivariate logistic regression analysis, addition of MV TF PCA conferred diagnostic benefit to the single variables, CA-125 (p=0.052) and D-dimer (p=0.019). Perioperative VTE occurred in 16% of cancer patients and was associated with an advanced FIGO stage, but not MV TF PCA. There was no difference in plasma TF antigen levels between study groups. CONCLUSIONS: MV TF PCA, but not plasma TF antigen, may provide valuable additional information for the diagnostic work-up of women with suspected ovarian cancer.


Subject(s)
Cell-Derived Microparticles/pathology , Ovarian Neoplasms/complications , Ovarian Neoplasms/diagnosis , Thromboplastin/analysis , Venous Thromboembolism/complications , Venous Thromboembolism/diagnosis , Adult , Aged , Biomarkers, Tumor/analysis , Biomarkers, Tumor/blood , CA-125 Antigen/analysis , CA-125 Antigen/blood , Female , Fibrin Fibrinogen Degradation Products/analysis , Hemostasis , Humans , Middle Aged , Ovarian Neoplasms/blood , Ovarian Neoplasms/pathology , Ovary/pathology , P-Selectin/blood , Perioperative Period , Preoperative Period , Venous Thromboembolism/blood , Venous Thromboembolism/pathology
6.
Blood ; 126(19): 2230-8, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26396093

ABSTRACT

The CD32a immunoglobulin G (IgG) receptor (Fcγ receptor IIa) is a potential therapeutic target for diseases in which IgG immune complexes (ICs) mediate inflammation, such as heparin-induced thrombocytopenia, rheumatoid arthritis, and systemic lupus erythematosus. Monoclonal antibodies (mAbs) are a promising strategy for treating such diseases. However, IV.3, perhaps the best characterized CD32a-blocking mAb, was recently shown to induce anaphylaxis in immunocompromised "3KO" mice. This anaphylactic reaction required a human CD32a transgene because mice lack an equivalent of this gene. The finding that IV.3 induces anaphylaxis in CD32a-transgenic mice was surprising because IV.3 had long been thought to lack the intrinsic capacity to trigger cellular activation via CD32a. Such an anaphylactic reaction would also limit potential therapeutic applications of IV.3. In the present study, we examine the molecular mechanisms by which IV.3 induces anaphylaxis. We now report that IV.3 induces anaphylaxis in immunocompetent CD32a-transgenic "FCGR2A" mice, along with the novel finding that IV.3 and 2 other well-characterized CD32a-blocking mAbs, AT-10 and MDE-8, also induce severe thrombocytopenia in FCGR2A mice. Using recombinant variants of these same mAbs, we show that IgG "Fc" effector function is necessary for the induction of anaphylaxis and thrombocytopenia in FCGR2A mice. Variants of these mAbs lacking the capacity to activate mouse IgG receptors not only failed to induce anaphylaxis or thrombocytopenia, but also very potently protected FCGR2A mice from near lethal doses of IgG ICs. Our findings show that effector-deficient IV.3, AT-10, and MDE-8 are promising candidates for developing therapeutic mAbs to treat CD32a-mediated diseases.


Subject(s)
Anaphylaxis/chemically induced , Antibodies, Monoclonal, Murine-Derived/adverse effects , Antibodies, Neutralizing/adverse effects , Receptors, IgG/antagonists & inhibitors , Thrombocytopenia/chemically induced , Anaphylaxis/immunology , Anaphylaxis/pathology , Animals , Antibodies, Monoclonal, Murine-Derived/immunology , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , Humans , Mice , Mice, Transgenic , Receptors, IgG/genetics , Receptors, IgG/immunology , Thrombocytopenia/genetics , Thrombocytopenia/immunology , Thrombocytopenia/pathology
7.
Exp Hematol Oncol ; 4: 22, 2015.
Article in English | MEDLINE | ID: mdl-26251762

ABSTRACT

BACKGROUND: In acute myeloid leukemia (AML), disseminated intravascular coagulation (DIC) contributes to morbidity and mortality, but the underlying pathomechanisms remain incompletely understood. METHODS: We conducted a prospective study on 69 patients with newly diagnosed AML to further define the correlates of systemic coagulation activation in this hematological malignancy. Tissue factor procoagulant activity (TF PCA) of isolated peripheral blood mononuclear cells (PBMCs) and TF expression by circulating microparticles (MPs) were assessed by single-stage clotting and thrombin generation assay, respectively. Soluble plasma TF antigen and secretion of vascular endothelial growth factor (VEGF) by cultured PBMCs were measured by ELISA. Cell-free plasma DNA was quantified by staining with a fluorescent dye. RESULT: TF PCA of PBMCs was significantly increased in AML patients as compared to healthy controls. Furthermore, TF PCA was significantly associated with decompensated DIC at presentation, as defined by a plasma fibrinogen level of ≤1 g/L (n = 11). In addition to TF PCA and circulating blasts, serum lactate dehydrogenase, a surrogate marker for leukemic cell turnover, correlated with plasma D-Dimer in the total patient cohort and was significantly increased in DIC patients, suggesting a role for myeloblast apoptosis/necrosis in activation of the TF-dependent coagulation pathway. Consistently, TF-bearing plasma MPs were more frequently detected and levels of soluble TF antigen were significantly higher in DIC vs. non-DIC patients. No association was found between TF PCA expression and VEGF secretion by isolated PBMCs, but significantly increased levels of cell-free plasma DNA pointed to a contribution of the intrinsic contact pathway to systemic coagulation activation in the total patient cohort and in patients with lower TF PCA expression. While PBMC-associated TF PCA had no effect on long-term survival, DIC occurrence at presentation increased the risk of early mortality. CONCLUSION: In newly diagnosed AML, TF expression by PBMCs and shedding of TF-bearing plasma MPs are central to the pathogenesis of DIC, but additional pathways, such as DNA liberation, may contribute to systemic coagulation activation.

8.
Blood Coagul Fibrinolysis ; 24(5): 510-7, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23406663

ABSTRACT

Preoperative evaluation of patients presenting with ovarian masses is challenging, partly due to shortcomings with the commonly used marker, CA-125. Ovarian cancer is associated with systemic coagulation activation. Measurement of D-dimer, serum tissue factor (TF), and the coagulation process as a whole are considered candidates for improving discrimination between benign and malignant ovarian masses. We therefore sought to identify possible benefits by analyzing preoperative coagulation status in conjunction with CA-125 in patients with ovarian masses. Preoperative blood from 95 patients with ovarian masses (75 benign, 20 malignant) and 30 controls was analyzed, prospectively. Thromboelastography served for global hemostatic assessment. Plasma TF antigen and D-dimer were measured by ELISA and microparticle-associated TF activity by thrombin generation assay. TF microparticles were enumerated by flow cytometry. Time to clot formation by thromboelastography was similar between patients having either benign or malignant ovarian tumors. Clot formation rate, clot strength, and coagulation index were significantly increased in patients having malignant versus benign tumors, indicating that thromboelastography differentiated malignant from benign tumors. D-dimer alone differentiated malignant from benign ovarian tumors and also improved differentiation when combined with CA-125. Circulating TF antigen, activity, and TF microparticle numbers, however, failed to differentiate benign from malignant tumors. Significant coagulation activation occurs in women with ovarian malignancies. Plasma D-dimer may help discriminate between patients with benign and malignant tumors. Thromboelastography may also contribute meaningfully when combined with CA-125 in the preoperative evaluation of ovarian masses. Larger studies are needed to assess these possibilities.


Subject(s)
Blood Coagulation/physiology , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/physiopathology , Adult , Aged , Biomarkers, Tumor/blood , CA-125 Antigen/blood , Diagnosis, Differential , Female , Fibrin Fibrinogen Degradation Products , Fibrinogen/analysis , Hemostasis , Humans , Middle Aged , Preoperative Period , Sensitivity and Specificity , Thrombelastography , Thromboplastin/analysis
9.
Blood ; 119(13): 3164-75, 2012 Mar 29.
Article in English | MEDLINE | ID: mdl-22327225

ABSTRACT

Tissue factor (TF) expression by tumor cells correlates with metastasis clinically and supports metastasis in experimental settings. However, the precise pathways coupling TF to malignancy remain incompletely defined. Here, we show that clot formation by TF indirectly enhances tumor cell survival after arrest in the lung, during experimental lung metastasis, by recruiting macrophages characterized by CD11b, CD68, F4/80, and CX(3)CR1 (but not CD11c) expression. Genetic or pharmacologic inhibition of coagulation, by either induction of TF pathway inhibitor ex-pression or by treatment with hirudin, respectively, abrogated macrophage recruitment and tumor cell survival. Furthermore, impairment of macrophage function, in either Mac1-deficient mice or in CD11b-diphtheria toxin receptor mice in which CD11b-positive cells were ablated, decreased tumor cell survival without altering clot formation, demonstrating that the recruitment of functional macrophages was essential for tumor cell survival. This effect was independent of NK cells. Moreover, a similar population of macrophages was also recruited to the lung during the formation of a premetastatic niche. Anticoagulation inhibited their accumulation and prevented the enhanced metastasis associated with the formation of the niche. Our study, for the first time, links TF induced coagulation to macrophage recruitment in the metastatic process.


Subject(s)
Blood Coagulation/drug effects , Cell Movement/drug effects , Macrophages/drug effects , Monocytes/drug effects , Neoplasms/pathology , Stem Cell Niche/physiology , Thromboplastin/pharmacology , Animals , Blood Coagulation/physiology , Cell Movement/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Humans , Macrophages/metabolism , Macrophages/physiology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, SCID , Mice, Transgenic , Monocytes/metabolism , Monocytes/physiology , Neoplasm Metastasis , Neoplasms/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/physiology , Stem Cell Niche/drug effects , Thromboplastin/metabolism
12.
Platelets ; 21(7): 571-4, 2010.
Article in English | MEDLINE | ID: mdl-20849210

ABSTRACT

The multifunctional cytokine, TWEAK (TNF-like weak inducer of apoptosis), is a member of the TNFα superfamily. TWEAK is found in a broad range of cell types and has been linked to cell growth and survival, angiogenesis and other inflammatory processes. These functions and their importance in inflammatory diseases have made TWEAK an attractive pharmaceutical target, particularly for immunotherapy with monoclonal antibodies (mAbs). Immunotherapy targeting another TNFα family member, CD154, was associated with thrombosis in clinical trials. Subsequent studies identified platelets, which contain CD154, as a possible contributing factor to thrombosis in these trials. Since clinical trials with anti-TWEAK mAbs have already begun, we considered it important to determine whether platelets contain TWEAK. Using a variety of immunologic methods we found that, upon activation, human platelets expose TWEAK antigen and release it in soluble form (sTWEAK). By flow cytometry we determined that human platelets activated by TRAP (Thrombin Receptor Agonist Peptide) and other agonists expose TWEAK antigen (22% median positivity) and release TWEAK positive microparticles. The presence of TWEAK on platelets was confirmed by confocal microscopy. By ELISA, we found that sTWEAK is released by activated platelets. Finally, western blot analysis revealed TWEAK protein (34 kDa) in washed platelet lysates. The finding that human platelets contain TWEAK raises important questions about its possible functions in normal physiology, as well as in inflammatory diseases and their treatment.


Subject(s)
Blood Platelets/metabolism , Receptors, Tumor Necrosis Factor/blood , Flow Cytometry , Humans , TWEAK Receptor
13.
Methods Mol Biol ; 663: 241-59, 2010.
Article in English | MEDLINE | ID: mdl-20617422

ABSTRACT

It is well established that the blood coagulation system is activated in cancer. In addition, there is considerable evidence to suggest that clotting activation plays an important role in the biology of malignant tumors, including the process of blood-borne metastasis. For many years our laboratory has used experimental models of lung metastasis to study the events that follow the introduction of procoagulant-bearing tumor cells into circulating blood. This chapter focuses on the basic methods involved in assessing the anti-metastatic effects of anticoagulants and anti-platelet agents using rodent models of experimental metastasis. In addition, it summarizes our experience with these models, which collectively suggests that intravascular coagulation and platelet activation are a necessary prelude to lung tumor formation and that interruption of coagulation pathways or platelet aggregation may be an effective anti-metastatic strategy.


Subject(s)
Anticoagulants/pharmacology , Antineoplastic Agents/pharmacology , Lung Neoplasms/pathology , Neoplasm Metastasis/drug therapy , Platelet Aggregation Inhibitors/pharmacology , Animals , Anticoagulants/therapeutic use , Antineoplastic Agents/therapeutic use , Blood Coagulation/drug effects , Cell Line, Tumor , Enoxaparin/pharmacology , Enoxaparin/therapeutic use , Humans , Lung Neoplasms/drug therapy , Mice , Neoplasms, Experimental , Platelet Aggregation Inhibitors/therapeutic use , Thromboplastin/antagonists & inhibitors , Warfarin/pharmacology , Warfarin/therapeutic use
14.
J Immunol ; 185(3): 1577-83, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20585032

ABSTRACT

Anti-CD40L immunotherapy in systemic lupus erythematosus patients was associated with thromboembolism of unknown cause. We previously showed that monoclonal anti-CD40L immune complexes (ICs) activated platelets in vitro via the IgG receptor (FcgammaRIIa). In this study, we examined the prothrombotic effects of anti-CD40L ICs in vivo. Because mouse platelets lack FcgammaRIIa, we used FCGR2A transgenic mice. FCGR2A mice were injected i.v. with preformed ICs consisting of either anti-human CD40L mAb (M90) plus human CD40L, or a chimerized anti-mouse CD40L mAb (hMR1) plus mouse CD40L. ICs containing an aglycosylated form of hMR1, which does not bind FcgammaRIIa, were also injected. M90 IC caused shock and thrombocytopenia in FCGR2A but not in wild-type mice. Animals injected with hMR1 IC also experienced these effects, whereas those injected with aglycosylated-hMR1 IC did not, demonstrating that anti-CD40L IC-induced platelet activation in vivo is FcgammaRIIa-dependent. Sequential injections of individual IC components caused similar effects, suggesting that ICs were able to assemble in circulation. Analysis of IC-injected mice revealed pulmonary thrombi consisting of platelet aggregates and fibrin. Mice pretreated with a thrombin inhibitor became moderately thrombocytopenic in response to anti-CD40L ICs and had pulmonary platelet-thrombi devoid of fibrin. In conclusion, we have shown for the first time that anti-CD40L IC-induced thrombosis can be replicated in mice transgenic for FcgammaRIIa. This molecular mechanism may be important for understanding thrombosis associated with CD40L immunotherapy. The FCGR2A mouse model may also be useful for assessing the hemostatic safety of other therapeutic Abs.


Subject(s)
Antigen-Antibody Complex/physiology , Autoantibodies/toxicity , CD40 Ligand/immunology , Platelet Activation/immunology , Receptors, IgG/genetics , Thrombosis/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/toxicity , Antigen-Antibody Complex/administration & dosage , Antigen-Antibody Complex/toxicity , Autoantibodies/administration & dosage , Autoantibodies/therapeutic use , Humans , Hybridomas , Mice , Mice, Knockout , Mice, Transgenic , Platelet Activation/genetics , Receptors, IgG/deficiency , Receptors, IgG/physiology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/physiology , Recombinant Fusion Proteins/toxicity , Thrombosis/blood
15.
J Thromb Thrombolysis ; 30(2): 142-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-19911252

ABSTRACT

Heparin-platelet factor 4 (PF4) antibodies mediate heparin-induced thrombocytopenia (HIT) and, irrespective of thrombocytopenia, are associated with poorer outcomes in some patients. The prevalence of heparin-PF4 antibodies, including platelet-activating ones, in patients in the medical, neurotrauma, or shock-trauma intensive care unit (ICU) remains unclear. In this single-center, observational study, heparin-PF4 antibodies (IgG/A/M) were measured by ELISA in 185 adults (median APACHE II score, 16) admitted to the medical (n = 27), neurotrauma (n = 96), or shock-trauma (n = 62) ICU and after 7 +/- 2 days. Seropositive patients and heparin-treated patients with unexplained, new-onset thrombocytopenia were also tested for platelet-activating antibodies using a serotonin release assay (SRA). Of 185 patients, seropositivity occurred in 20 patients (10.8%; 95% CI 6.7-16.2%) at admission and 54 (29.2%, 95% CI 22.8-36.3%) after 7 days (P < 0.001). Platelet-activating antibodies occurred in 4 seropositive patients at admission and 9 seropositive patients after 7 days (including in 1 patient at each assessment), each without thrombocytopenia or new thrombosis. Of 12 seropositive patients with platelet-activating antibodies, 6 had an ELISA optical density (OD) >1.0. ELISA-positive, SRA-negative, suspected HIT occurred in 1 patient. Heparin-PF4 antibodies are present in 10.8% of medical, neurotrauma, or shock-trauma ICU patients at admission and increase significantly to 29.2% within 7 days. Approximately 17-20% of seropositive ICU patients, often those with an ELISA OD >1.0, have platelet-activating heparin-PF4 antibodies.


Subject(s)
Antibodies/blood , Anticoagulants/immunology , Critical Care , Heparin/immunology , Intensive Care Units , Platelet Factor 4/immunology , Thrombocytopenia/immunology , APACHE , Enzyme-Linked Immunosorbent Assay , Female , Hospital Mortality , Humans , Length of Stay , Male , Middle Aged , Platelet Activation/immunology , Prospective Studies , Seroepidemiologic Studies , Texas/epidemiology , Thrombocytopenia/blood , Thrombocytopenia/epidemiology , Thrombocytopenia/mortality , Time Factors , Treatment Outcome
16.
Clin Exp Metastasis ; 26(7): 829-37, 2009.
Article in English | MEDLINE | ID: mdl-19642003

ABSTRACT

It is established that experimental metastasis requires platelet activity. CD154 expressed on and released from activated platelets induces an inflammatory response in endothelial cells and monocytes, including tissue factor production. CD154 has also been shown to activate platelets in vitro and promote thrombus stability in vivo. These CD154 effects may be mediated, at least in part, by CD40 signaling on platelets and vascular endothelial cells. We have previously demonstrated prolonged bleeding and PFA-100 closure times in mice deficient for Cd154 or its receptor Cd40. In the present study, we hypothesized that Cd40 and Cd154 promote lung tumor formation in experimental metastasis in mice. We created mice doubly deficient in Cd40 and Cd154 (Dbl KO) and found them to be both fertile and viable. Injected tumor cells seeded poorly in mice deficient in Cd40 or Cd154, as well as Dbl KO, compared to wild-type mice. We sought to determine whether blood-borne Cd40 versus endothelial Cd40 contribute differentially to reduced experimental lung metastasis, as observed in Cd40 deficient mice. By bone marrow transplantation, we created mice deficient for Cd40 either in the blood compartment but not in the endothelium, or vice versa. We found that mice deficient in blood compartment Cd40 had fewer lung nodules compared to wild-type mice and mice deficient in endothelial Cd40. Our findings suggest an important contribution of the Cd40-Cd154 pathway to experimental lung metastasis. Furthermore, the data points to a selective role for peripheral blood cell Cd40 in this process.


Subject(s)
CD40 Antigens/immunology , CD40 Ligand/immunology , Lung Neoplasms/secondary , Melanoma, Experimental/pathology , Animals , Bone Marrow Transplantation , Cell Line, Tumor , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Melanoma, Experimental/immunology , Mice , Mice, Knockout , Neoplasm Metastasis
17.
Thromb Haemost ; 101(6): 1147-55, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19492160

ABSTRACT

Tissue factor (TF) plays a critical role in tumour growth and metastasis, and its enhanced release into plasma in association with cellular microparticles (MPs) has recently been associated with pathological cancer progression. We have previously demonstrated significantly elevated levels of plasma TF antigen as well as systemic coagulation and platelet activation in patients with localised prostate cancer. In this prospective study, we used a highly sensitive one-stage clotting assay to measure preoperative TF-specific procoagulant activity (PCA) of plasma MPs in 68 consecutive patients with early-stage prostate cancer to further explore the relevance of circulating TF in this tumour entity. Automated calibrated thrombography was used to monitor thrombin generation in cell-free plasma samples in the absence of exogenous TF or phospholipids. Compared to healthy male controls (n=20), patients had significantly increased levels of both D-dimer and TF-specific PCA of plasma MPs (p<0.001). Furthermore, MP-associated TF PCA was higher in patients with (n=29) than in those without (n=39) laboratory evidence of an acute-phase reaction (p=0.004) and decreased to normal levels within one week after radical prostatectomy. Overall, we found a significant correlation between TF-specific PCA of plasma MPs and plasma D-dimer (p=0.002), suggesting that plasma MPs contributed to in-vivo coagulation activation in a TF-dependent manner. Thrombin generation in plasma was also significantly increased in patients compared to controls (p<0.01). Collectively, our findings suggest that TF-specific PCA of plasma MPs contributes to intravascular coagulation activation in patients with early-stage prostate cancer and may represent a potential link between hypercoagulability, inflammation, and disease progression.


Subject(s)
Acute-Phase Reaction/physiopathology , Cell-Derived Microparticles/metabolism , Prostatic Neoplasms/physiopathology , Thrombin/metabolism , Thromboplastin/metabolism , Acute-Phase Reaction/diagnosis , Acute-Phase Reaction/etiology , Acute-Phase Reaction/pathology , Aged , Antibodies, Monoclonal , Blood Coagulation , Blood Coagulation Tests , Cell Line, Tumor , Cell Separation , Disease Progression , Flow Cytometry , Humans , Lipopolysaccharides/metabolism , Male , Middle Aged , Neoplasm Staging , Prospective Studies , Prostatic Neoplasms/complications , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/pathology , Sensitivity and Specificity , Thrombin/genetics , Thromboplastin/immunology
18.
Semin Thromb Hemost ; 33(7): 643-52, 2007 Oct.
Article in English | MEDLINE | ID: mdl-18000790

ABSTRACT

Clotting activation occurs frequently in cancer. Tissue factor (TF), the most potent initiator of coagulation, is expressed aberrantly in many types of malignancy and is involved not only in tumor-associated hypercoagulability but also in promoting tumor angiogenesis and metastasis via coagulation-dependent and coagulation-independent (signaling) mechanisms. Tissue factor pathway inhibitor (TFPI) is the natural inhibitor of TF coagulant and signaling activities. Studies have shown that TFPI exhibits antiangiogenic and antimetastatic effects in vitro and in vivo. In animal models of experimental metastasis, both circulating and tumor cell-associated TFPI are shown to significantly reduce tumor cell-induced coagulation activation and lung metastasis. Heparins and heparin derivatives, which induce the release of TFPI from the vascular endothelium, also exhibit antitumor effects, and TFPI may contribute significantly to those effects. Indeed, a non-anticoagulant low-molecular-weight heparin with intact TFPI-releasing capacity has been shown to have significant antimetastatic effect in a similar experimental mouse model. The evidence supporting the dual inhibitory functions on TF-driven coagulation and signaling strengthen the rationale for considering TFPI as a potential anticancer agent. This article primarily summarizes the evidence for antiangiogenic and antimetastatic effects of TFPI and describes its potential mechanisms of action. The possible application of TFPI and other inhibitors of TF as potential anticancer agents is described, and information regarding potential antitumor properties of TFPI-2 (which has structural similarities to TFPI) is also included.


Subject(s)
Hemostasis , Lipoproteins/metabolism , Lipoproteins/pharmacology , Neoplasm Metastasis , Neoplasms/blood , Neoplasms/pathology , Thromboplastin/antagonists & inhibitors , Animals , Anticoagulants/metabolism , Anticoagulants/pharmacology , Fibrinolytic Agents/metabolism , Fibrinolytic Agents/pharmacology , Heparin, Low-Molecular-Weight/metabolism , Heparin, Low-Molecular-Weight/pharmacology , Humans , Neoplasm Metastasis/prevention & control , Neoplasms/blood supply , Neovascularization, Pathologic , Serine Proteinase Inhibitors/metabolism , Serine Proteinase Inhibitors/pharmacology , Thromboplastin/metabolism
19.
Am J Emerg Med ; 25(3): 279-84, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17349901

ABSTRACT

BACKGROUND: Patients with heparin-platelet factor 4 (PF4) antibodies, particularly platelet-activating ones, are at risk for heparin-induced thrombocytopenia if administered heparin. We determined the heparin-PF4 antibody prevalence in emergency department (ED) patients presenting with chest pain or symptoms of thrombosis. METHODS: Admission samples from 324 ED patients with chest pain or symptoms of thrombosis were tested for heparin-PF4 antibodies and, if positive, platelet-activating antibodies. RESULTS: Twenty-four (7.4%; 95% confidence interval, 4.8%-10.8%) patients had heparin-PF4 antibodies. Seropositivity occurred in 18 (9.2%) of 196 patients recently (< or =6 months) hospitalized vs 6 (4.7%) of 128 not recently hospitalized (P = .19), and in 16/231 (6.9%) patients with chest pain vs 8/93 (8.6%) with other thrombosis (P = .64). Of 22 seropositive patients retested, 8 (7 recently hospitalized) had platelet-activating antibodies. CONCLUSION: Heparin-PF4 antibody prevalence is 7.4% in ED patients with chest pain or thrombosis, with approximately 1 in 3 seropositive patients having platelet-activating antibodies. Alternative, nonheparin anticoagulation would be prudent in these at-risk patients.


Subject(s)
Antibodies/blood , Anticoagulants/adverse effects , Emergency Service, Hospital/statistics & numerical data , Heparin/adverse effects , Platelet Factor 4/immunology , Thrombocytopenia/chemically induced , Thrombosis/immunology , Female , Humans , Male , Middle Aged , Platelet Factor 4/drug effects , Prospective Studies , Thrombocytopenia/immunology , Thrombosis/blood
20.
Thromb Haemost ; 97(3): 464-70, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17334515

ABSTRACT

Tissue factor (TF) is involved in cancer growth and metastasis, and haemostatic abnormalities are found in most patients with advanced malignancies, including prostate cancer (PC). Because anti-haemostatic agents are increasingly screened for their potential to prolong survival in tumor patients, a detailed characterization of haemostatic markers in selected cancer subtypes and clinical stages is warranted. In this study, we measured preoperative plasma TF antigen in a large cohort of patients with localized PC and correlated its levels with markers of coagulation and platelet activation, prostate-specific antigen (PSA), and histopathological findings to explore its potential as a prognostic marker in this tumor entity. Out of 140 patients, 19% and 23% had plasma TF antigen levels of <40 pg/ml (low-TF) and >200 pg/ml (high-TF), respectively, which was substantially higher than in 42 healthy male controls. Patients also had low-grade systemic coagulation activation as evidenced by elevated D-dimer, F1 + 2, and PAP plasma levels. Furthermore, similar to sP-selectin and sCD40L antigen, flow cytometric analysis of platelet-derived microparticles in plasma revealed significantly increased numbers in high-TF as compared to low-TF patients and controls. Whereas elevated D-dimer was associated with larger and less differentiated tumors, preoperative plasma TF antigen levels (median [IQR]) were higher in patients with (161 pg/ml [100-236]) than in those without recurrent PC (105 pg/ml [52-182]), as indicated by a serum PSA of >0.1 ng/ml during ambulatory follow-up. In patients with localized PC, preoperative plasma TF antigen levels correlate with platelet activation in vivo and may indicate an increased risk for recurrent disease.


Subject(s)
Antigens, Neoplasm/blood , Biomarkers, Tumor/blood , Blood Coagulation , Platelet Activation , Prostatic Neoplasms/blood , Thromboplastin/metabolism , CD40 Ligand/blood , Cell Differentiation , Disseminated Intravascular Coagulation/blood , Enzyme-Linked Immunosorbent Assay , Fibrin Fibrinogen Degradation Products/metabolism , Fibrinolysin/metabolism , Flow Cytometry , Follow-Up Studies , Hemostasis , Humans , Male , Middle Aged , Neoplasm Invasiveness , P-Selectin/blood , Peptide Fragments/blood , Prognosis , Prospective Studies , Prostate-Specific Antigen/blood , Prostatectomy , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Prothrombin , Recurrence , Risk Assessment , Time Factors , alpha-2-Antiplasmin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...