Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
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
2.
Angiogenesis ; 19(1): 79-94, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26650228

ABSTRACT

Human tear lipocalin (Tlc) was utilized as a protein scaffold to engineer an Anticalin that specifically binds and functionally blocks vascular endothelial growth factor A (VEGF-A), a pivotal inducer of physiological angiogenesis that also plays a crucial role in several neovascular diseases. Starting from a naive combinatorial library where residues that form the natural ligand-binding site of Tlc were randomized, followed by affinity maturation, the final Anticalin PRS-050 was selected to bind all major splice forms of VEGF-A with picomolar affinity. Moreover, this Anticalin cross-reacts with the murine ortholog. PRS-050 efficiently antagonizes the interaction between VEGF-A and its cellular receptors, and it inhibits VEGF-induced mitogenic signaling as well as proliferation of primary human endothelial cells with subnanomolar IC50 values. Intravitreal administration of the Anticalin suppressed VEGF-induced blood-retinal barrier breakdown in a rabbit model. To allow lasting systemic neutralization of VEGF-A in vivo, the plasma half-life of the Anticalin was extended by site-directed PEGylation. The modified Anticalin efficiently blocked VEGF-mediated vascular permeability as well as growth of tumor xenografts in nude mice, concomitantly with reduction in microvessel density. In contrast to bevacizumab, the Anticalin did not trigger platelet aggregation and thrombosis in human FcγRIIa transgenic mice, thus suggesting an improved safety profile. Since neutralization of VEGF-A activity is well known to exert beneficial effects in cancer and other neovascular diseases, including wet age-related macular degeneration, this Anticalin offers a novel potent small protein antagonist for differentiated therapeutic intervention in oncology and ophthalmology.


Subject(s)
Lipocalins/pharmacology , Molecular Targeted Therapy , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Antigen-Antibody Complex/metabolism , Bevacizumab/pharmacology , Bevacizumab/therapeutic use , Blood-Retinal Barrier/pathology , Capillary Permeability , Cell Proliferation/drug effects , Female , Half-Life , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Lipocalins/pharmacokinetics , Lipocalins/therapeutic use , Mice, Nude , Mice, Transgenic , Mitogens/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Polyethylene Glycols/chemistry , Protein Engineering , Rabbits , Receptors, IgG/metabolism , Signal Transduction , Surface Plasmon Resonance , Thrombosis/pathology , Vascular Endothelial Growth Factor A/metabolism
3.
Angiogenesis ; 19(3): 389-406, 2016 07.
Article in English | MEDLINE | ID: mdl-27234973

ABSTRACT

Anti-vascular endothelial growth factor (VEGF) therapies have improved clinical outcomes for patients with cancers and retinal vascular diseases. Three anti-VEGF agents, pegaptanib, ranibizumab, and aflibercept, are approved for ophthalmic indications, while bevacizumab is approved to treat colorectal, lung, and renal cancers, but is also used off-label to treat ocular vascular diseases. The efficacy of bevacizumab relative to ranibizumab in treating neovascular age-related macular degeneration has been assessed in several trials. However, questions persist regarding its safety, as bevacizumab can form large complexes with dimeric VEGF165, resulting in multimerization of the Fc domain and platelet activation. Here, we compare binding stoichiometry, Fcγ receptor affinity, platelet activation, and binding to epithelial and endothelial cells in vitro for bevacizumab and aflibercept, in the absence or presence of VEGF. In contrast to bevacizumab, aflibercept forms a homogenous 1:1 complex with each VEGF dimer. Unlike multimeric bevacizumab:VEGF complexes, the monomeric aflibercept:VEGF complex does not exhibit increased affinity for low-affinity Fcγ receptors, does not activate platelets, nor does it bind to the surface of epithelial or endothelial cells to a greater degree than unbound aflibercept or control Fc. The latter finding reflects the fact that aflibercept binds VEGF in a unique manner, distinct from antibodies not only blocking the amino acids necessary for VEGFR1/R2 binding but also occluding the heparin-binding site on VEGF165.


Subject(s)
Bevacizumab/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Recombinant Fusion Proteins/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/metabolism , Angiogenesis Inhibitors/therapeutic use , Animals , Antigen-Antibody Complex/chemistry , Antigen-Antibody Complex/metabolism , Bevacizumab/adverse effects , Bevacizumab/therapeutic use , Cell Line , Human Umbilical Vein Endothelial Cells , Humans , In Vitro Techniques , Macular Degeneration/immunology , Macular Degeneration/metabolism , Macular Degeneration/therapy , Mice , Mice, Transgenic , Platelet Activation , Protein Binding , Protein Multimerization , Receptors, IgG/genetics , Receptors, IgG/metabolism , Receptors, Vascular Endothelial Growth Factor/therapeutic use , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/therapeutic use , Thrombocytopenia/etiology , Thrombosis/etiology , Vascular Endothelial Growth Factor A/immunology
4.
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
5.
J Cardiovasc Nurs ; 29(2): 130-9, 2014.
Article in English | MEDLINE | ID: mdl-23364574

ABSTRACT

BACKGROUND: Women with systemic lupus erythematosus (SLE) display a 7- to 10-fold increased risk for cardiovascular disease (CVD) compared with non-SLE controls, yet many are unaware of this risk despite years spent in the healthcare system. It is not clear why they lack awareness of increased CVD risk or which factors influence awareness. OBJECTIVE: The purpose of this study was to assess in women with SLE their perceived CVD risk, the association between clinically identified and perceived CVD risk factors, and factors that influenced CVD risk awareness and adoption of risk-reducing behaviors. METHODS: Questionnaires, face-to-face meetings, and clinical assessments were used to collect data on demographics, perceived CVD risk, perceived CVD risk factors, actual CVD risk factors, risk-reducing behaviors, and healthcare provider counseling from 60 women with SLE. Regression analyses identified factors that influenced risk awareness and adoption of risk-reducing behaviors. RESULTS: Two-thirds of the participants perceived themselves at increased CVD risk when compared with women without SLE, but the same number did not perceive an increase in their absolute CVD risk. Age was a significant predictor (P = .05) for awareness of increased absolute risk; younger age correlated with increased awareness. Most women received information about heart disease from public media. On average, participants had 4 CVD risk factors but perceived that they had only 2. Age (P = .001) and the number of perceived risk factors (P = .004) predicted adoption of risk-reducing behaviors (P = .03). CONCLUSION: Participants underestimated their CVD risk factors and did not personalize their increased CVD risk. Healthcare providers' identification and discussion of CVD risk factors in women with SLE may enhance their CVD risk awareness and the adoption of risk-reducing behaviors.


Subject(s)
Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/prevention & control , Health Knowledge, Attitudes, Practice , Lupus Erythematosus, Systemic/epidemiology , Risk Reduction Behavior , Aged , Communication , Comorbidity , Female , Humans , Patient Education as Topic , Physician-Patient Relations , Risk Assessment , Risk Factors
6.
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
7.
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
8.
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
9.
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
10.
Thromb Haemost ; 96(6): 816-21, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17139378

ABSTRACT

Low-molecular-weight heparins (LMWH) exhibit potent anticoagulant efficacy via their plasmatic effects on thrombin and factor Xa. These agents are also effective in releasing endothelial tissue factor pathway inhibitor (TFPI), the natural inhibitor of tissue factor, and exhibit significant anti-metastatic effects in experimental animal models. However, the potential for bleeding complications has slowed down the more widespread adoption of LMWH therapy in cancer patients. In this study, the effect of a non-anticoagulant form of LMWH (NA-LMWH) on experimental lung metastasis and tumor cell-induced platelet aggregation in vivo was compared to the LMWH enoxaparin. Using the B16 melanoma mouse model of metastasis, subcutaneous (s.c.) injection of NA-LMWH or enoxaparin (10 mg/kg), three hours before intravenous (i.v.) injection of metastatic melanoma cells, followed by daily doses for 14 days, reduced lung tumor formation by 70% (P < 0.001). I.v. injection of tumor cells resulted in a significant (50-62%, P < 0.01) fall in platelet counts. Pre-injection (i.v.) of enoxaparin completely abolished the tumor cell-induced thrombocytopenia, whereas NA-LMWH had no effect. Four hours after a single s.c. dose, enoxaparin but not NA-LMWH prolonged the clotting time three-fold and delayed the time to clot initiation more than 10-fold as measured by a Sonoclot analyzer and by thromboelastography, respectively. Enoxaparin but not NA-LMWH demonstrated a significant anticoagulant effect in mice. Both NA-LMWH and enoxaparin caused similar TFPI release from endothelial cells in vitro. These data provide evidence to support the potential of NA-LMWH as an anti-metastatic agent without any significant impact on coagulation.


Subject(s)
Anticoagulants/pharmacology , Antineoplastic Agents/pharmacology , Enoxaparin/pharmacology , Heparin, Low-Molecular-Weight/analogs & derivatives , Lung Neoplasms/prevention & control , Melanoma, Experimental/drug therapy , Animals , Anticoagulants/therapeutic use , Antineoplastic Agents/therapeutic use , Blood Coagulation/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Enoxaparin/therapeutic use , Factor Xa Inhibitors , Heparin, Low-Molecular-Weight/pharmacology , Heparin, Low-Molecular-Weight/therapeutic use , Lipoproteins/metabolism , Lung Neoplasms/blood , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Melanoma, Experimental/blood , Melanoma, Experimental/complications , Melanoma, Experimental/pathology , Mice , Partial Thromboplastin Time , Platelet Aggregation/drug effects , Platelet Count , Prothrombin/antagonists & inhibitors , Thrombelastography , Thrombocytopenia/etiology , Thrombocytopenia/prevention & control
11.
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
12.
Thromb Haemost ; 94(5): 1084-93, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16363253

ABSTRACT

Clinical and experimental evidence suggests that the blood coagulation system is involved in the dissemination of malignant tumors. Consequently, anticoagulant agents have been tested as metastasis suppressors in experimental models. Recently, we have found a close correlation between factor Xa (FXa)-specificity of a series of synthetic serine protease inhibitors and their anti-metastatic potential in a murine T-cell lymphoma metastasis model. Interference of such inhibitors with blood-coagulation may represent a major experimental and clinical obstacle. Here, we test anti-metastatic effects of a recently developed, highly specific 3-amidinophenylalanine-type FXa inhibitor, WX-FX4, with weaker anticoagulant activity when compared to well-established FXa inhibitors, such as DX-9065a, as measured by the activated partial thromboplastin time, prothrombin time, prothrombinase complex activity, and coagulation time. Treatment of mice with WX-FX4 (1.5 mg/kg twice daily) led to significant reduction of experimental liver metastasis of a syngeneic T-cell lymphoma in DBA/2 mice (> 90%), and of experimental lung metastasis of a human fibrosarcoma in CD1 nu/nu mice (> 60%). Due to its relatively low anticoagulant activity, daily treatment over 100 days was possible, leading to significant survival benefits without inducing bleeding anomalities. FXa-inhibitors with highly efficient anti-metastatic potential without coagulation-related side effects may represent important new tools as anticancer agents.


Subject(s)
Adamantane/analogs & derivatives , Anticoagulants/pharmacology , Factor Xa Inhibitors , Lymphatic Metastasis , Lymphoma, T-Cell/drug therapy , Serine Proteinase Inhibitors/pharmacology , Adamantane/chemistry , Adamantane/pharmacology , Animals , Anticoagulants/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Factor Xa/metabolism , Female , Fibrosarcoma/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/secondary , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Mice , Mice, Inbred DBA , Naphthalenes/pharmacology , Propionates/pharmacology , Serine Proteinase Inhibitors/chemistry , Specific Pathogen-Free Organisms , Whole Blood Coagulation Time
13.
Thromb Haemost ; 93(6): 1137-46, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15968400

ABSTRACT

Our initial finding that CD40- and CD40 ligand (CD40L)-deficient mice displayed prolonged tail bleeding and platelet function analyzer (PFA-100) closure times prompted us to further investigate the role of the CD40-CD40L dyad in primary hemostasis and platelet function. Recombinant human soluble CD40L (rhsCD40L), chemical cross-linking of which suggested a trimeric structure of the protein in solution, activated platelets in a CD40-dependent manner as evidenced by increased CD62P expression. CD40 monoclonal antibody (mAb) M3, which completely blocked rhsCD40L-induced platelet activation, also prolonged PFA-100 closure times of normal human blood. In contrast, CD40 mAb G28-5 showed less potential in blocking rhsCD40L-induced CD62P expression and did not affect PFA-100 closure times. However, when added to the platelets after rhsCD40L, G28-5 significantly enhanced the platelet response by causing clustering of, and signaling through, FcgammaRII. Similarly, higher order multimeric immune complexes formed at a 1/3 molar ratio of M90, a CD40L mAb, to rhsCD40L induced strong Fcgamma RII-mediated platelet activation when translocated to the platelet surface in a CD40-dependent manner, including the induction of morphological shape changes, fibrinogen binding, platelet aggregation, dense granule release, microparticle generation and monocyte-platelet-conjugate formation. The results suggest that CD40 may play a role in primary hemostasis and platelet biology by two independent mechanisms: First, by functioning as a primary signaling receptor for CD40L and, second, by serving as a docking molecule for CD40L immune complexes. The latter would also provide a potential mechanistic explanation for the unexpected high incidence of CD40L mAb-associated thrombotic events in recent human and animal studies.


Subject(s)
CD40 Antigens/blood , CD40 Ligand/blood , Platelet Activation/immunology , Animals , Antibodies, Monoclonal/pharmacology , Antigen-Antibody Complex/blood , CD40 Antigens/immunology , CD40 Ligand/pharmacology , Hemostasis , Humans , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Activation/drug effects , Receptors, IgG/metabolism , Recombinant Proteins/pharmacology
14.
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.

15.
Arthritis Res Ther ; 17: 234, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26335795

ABSTRACT

INTRODUCTION: CD40 ligand (CD40L) blockade has demonstrated efficacy in experimental autoimmune models. However, clinical trials of hu5c8, an anti-human CD40L IgG1 antibody, in systemic lupus erythematosus (SLE) were halted due to an increased incidence of thrombotic events. This study evaluated CDP7657, a high affinity PEGylated monovalent Fab' anti-CD40L antibody fragment, to assess whether an Fc-deficient molecule retains efficacy while avoiding the increased risk of thrombotic events observed with hu5c8. METHODS: The potency and cross-reactivity of CDP7657 was assessed in in vitro assays employing human and non-human primate leukocytes, and the capacity of different antibody formats to activate platelets in vitro was assessed using aggregometry and dense granule release assays. Given the important role CD40L plays in regulating humoral immunity, in vivo efficacy was assessed by investigating the capacity of Cynomolgus monkeys to generate immune responses to the tetanus toxoid antigen while the potential to induce thrombotic events in vivo was evaluated after repeat dosing of antibodies to Rhesus monkeys. A PEGylated anti-mouse CD40L was generated to assess efficacy in the New Zealand Black/White (NZB/W) mouse model of SLE. RESULTS: CDP7657 dose-dependently inhibited antigen-specific immune responses to tetanus toxoid in Cynomolgus monkeys, and in contrast to hu5c8, there was no evidence of pulmonary thrombovasculopathy in Rhesus monkeys. Aglycosyl hu5c8, which lacks Fc receptor binding function, also failed to induce thrombotic events in Rhesus monkeys. In vitro experiments confirmed that antibody constructs lacking an Fc, including CDP7657, did not induce human or monkey platelet activation. A PEGylated monovalent Fab' anti-mouse CD40L antibody also inhibited disease activity in the NZB/W mouse model of SLE after administration using a therapeutic dosing regimen where mice received antibodies only after they had displayed severe proteinuria. CONCLUSIONS: These findings demonstrate for the first time that anti-CD40L antibodies lacking a functional Fc region do not induce thrombotic events in Rhesus monkeys and fail to activate platelets in vitro but, nevertheless retain pharmacological activity and support the investigation of CDP7657 as a potential therapy for systemic lupus erythematosus and other autoimmune diseases.


Subject(s)
Antibodies, Monoclonal/immunology , CD40 Ligand/immunology , Immunity, Humoral/immunology , Thrombosis/immunology , Animals , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , Antibody Formation/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/prevention & control , Disease Models, Animal , Humans , Immunity, Humoral/drug effects , Immunoglobulin Fab Fragments/immunology , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/prevention & control , Macaca fascicularis , Macaca mulatta , Mice, Inbred NZB , Polyethylene Glycols/chemistry , Tetanus Toxoid/immunology , Thrombosis/chemically induced
16.
Thromb Haemost ; 90(3): 549-54, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12958625

ABSTRACT

Platelets are known to play a role in blood borne metastasis. Previous experimental studies have suggested that platelet GpIIb/IIIa may be a therapeutic target. However, the need for intravenous administration limits the potential application of current GpIIb/IIIa inhibitors to cancer therapy. The aim of the present study was to assess the efficacy of a novel, non-peptide oral GpIIb/IIIa antagonist (XV454) on tumor cell-induced platelet aggregation in vivo and on experimental metastasis. A Lewis lung carcinoma (LL2) mouse model of experimental metastasis was used in this study. XV454 (100 micro g) was administered intravenously (via tail vein) or orally (gavages) to 20 g mice. To determine the effect of XV454 on platelet aggregation, blood samples were collected by cardiac puncture 10 minutes after intravenous and 1-24 hrs after oral XV454, and platelet function was assessed by aggregometry, thrombelastography and the Platelet Function Analyzer (PFA100). The effect of XV454 on tumor cell-induced thrombocytopenia was determined 10 minutes after intravenous and 3 hrs after oral XV454 administration. Tumor cells (2 x 10(6)) were injected intravenously and 15 minutes after cell injection, platelet count was measured and compared to baseline (pre-injection) counts. To assess the effect on metastasis, XV454 was administered intravenous or orally 10 minutes and 3 hrs before tumor cell injection, respectively. Eighteen days later, surface lung tumor nodules were counted and the total lung tumor burden assessed. In a fourth group, in addition to the initial oral dose (before tumor cell injection), oral XV454 was given daily for the first week and three times in the second week. Administration of XV454 (5 mg/kg) completely inhibited platelet aggregation and this effect persisted for at least 24 hrs after oral delivery. Both intravenous and oral XV454 significantly inhibited tumor cell-induced thrombocytopenia (P < 0.01), the number of surface lung tumor nodules (80-85%; P < 0.001) and total tumor burden (83% for intravenous group; 50% oral [single treatment] group; 91% oral [multiple treatment] group, P < 0.001). Overall, these data provide further evidence for the effect of oral and intravenous GpIIb/IIIa antagonism on tumor cell-platelet interaction and metastasis.


Subject(s)
Alanine/analogs & derivatives , Alanine/pharmacology , Carcinoma, Lewis Lung/blood , Lung Neoplasms/drug therapy , Neoplasm Metastasis/prevention & control , Oxazoles/pharmacology , Platelet Aggregation/drug effects , Administration, Oral , Alanine/administration & dosage , Animals , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Drug Evaluation, Preclinical , Female , Lung Neoplasms/blood , Lung Neoplasms/secondary , Mice , Mice, Nude , Neoplasm Metastasis/drug therapy , Oxazoles/administration & dosage , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Thrombocytopenia/etiology , Thrombocytopenia/prevention & control
17.
Thromb Haemost ; 87(6): 930-6, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12083498

ABSTRACT

The importance of tissue factor (TF) in tumor biology has been highlighted by studies suggesting its involvement in cell signaling, metastasis and angiogenesis. Since many animal studies have shown that anticoagulant therapy can reduce experimental metastasis, we studied whether the natural inhibitor of TF-mediated blood coagulation, Tissue Factor Pathway Inhibitor (TFPI), might be similarly effective. Using a murine experimental model, we found that intravenous injection of recombinant murine TFPI immediately before introduction of tumor cells reduced metastasis by 83% (P < 0.001). B16 murine melanoma cells stably transfected with a TFPI expression vector exhibited reduced lung seeding following intravenous injection by 81% (P < 0.001) compared with controls. No difference in primary tumor growth was observed between TFPI+ and control cells. Mice receiving intravenous somatic gene transfer of sense TFPI expression vector developed 78% fewer lung nodules than controls (P < 0.05). We conclude that TFPI has significant anti-metastatic activity in this experimental model.


Subject(s)
Anticoagulants/pharmacology , Lipoproteins/pharmacology , Lung Neoplasms/secondary , Melanoma, Experimental/pathology , Neoplasm Metastasis/prevention & control , Animals , Anticoagulants/administration & dosage , Endothelial Growth Factors/metabolism , Female , Heparin/administration & dosage , Heparin/pharmacology , Injections, Intravenous , Intercellular Signaling Peptides and Proteins/metabolism , Lipoproteins/administration & dosage , Lung Neoplasms/drug therapy , Lymphokines/drug effects , Lymphokines/metabolism , Melanoma, Experimental/complications , Melanoma, Experimental/drug therapy , Mice , Mice, Inbred C57BL , Neoplasm Metastasis/drug therapy , Platelet Count , Thrombocytopenia/drug therapy , Thrombocytopenia/etiology , Thrombocytopenia/prevention & control , Thrombophilia/drug therapy , Thrombophilia/etiology , Thrombophilia/prevention & control , Transfection , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
18.
Thromb Haemost ; 92(5): 1136-46, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15543344

ABSTRACT

AML patients may suffer from a disseminated coagulopathy, which can aggravate a pre-existing bleeding tendency due to thrombocytopenia and platelet dysfunction. The cellular and molecular mechanisms underlying this coagulopathy, however, are not completely understood. Indeed, the broad and increasing therapeutic use of cytotoxic drugs and growth factors is likely to contribute to the complexity of hemostatic abnormalities encountered in this hematologic malignancy. The nature of coagulation activation in AML was therefore investigated in vitro using the human leukemic cell line, HL60. Tissue factor (TF) was almost entirely located on the cell surface and bound factor VIIa, but only 15-25% of this TF was primarily functionally active. Treatment with increasing concentrations of daunorubicin or cytosine-beta-D-arabinofuranoside, two cytotoxic drugs commonly used in AML therapy, induced apoptosis and secondary necrosis of HL60 cells and resulted in marked decryption of TF PCA independent of de novo protein synthesis. This PCA-modulating effect was concomitant with and functionally dependent on the exposure of phosphatidylserine on the outer membrane leaflet. Similar observations were made in analogous ex vivo studies on patient-derived myeloblasts. Incubation of HL60 cells with GM-CSF, a cytokine expressed in the bone marrow microenvironment and used as an adjunct to AML treatment, evoked a cellular response, which included both enhanced TF production and release of VEGF-A and uPA into the culture medium. We conclude that both decryption of pre-formed TF PCA by chemotherapeutic drugs and de novo induction of TF by cytokines such as GM-CSF can regulate the pro-coagulant phenotype of HL60 cells in vitro.


Subject(s)
Antineoplastic Agents/pharmacology , Blood Coagulation , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Leukemia, Myeloid, Acute/blood , Thromboplastin/drug effects , Blood Cells/pathology , Cell Death/drug effects , Cells, Cultured , Cytarabine/pharmacology , Daunorubicin/pharmacology , Granulocyte Precursor Cells/drug effects , Granulocyte Precursor Cells/pathology , HL-60 Cells , Humans , Leukemia, Monocytic, Acute/blood , Leukemia, Monocytic, Acute/pathology , Leukemia, Myeloid, Acute/pathology , Leukemia, Myelomonocytic, Acute/blood , Leukemia, Myelomonocytic, Acute/pathology , Thromboplastin/biosynthesis , Thromboplastin/physiology
19.
Cancer Res ; 74(4): 1214-26, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24371232

ABSTRACT

Tissue factor (TF) is aberrantly expressed in solid cancers and is thought to contribute to disease progression through its procoagulant activity and its capacity to induce intracellular signaling in complex with factor VIIa (FVIIa). To explore the possibility of using tissue factor as a target for an antibody-drug conjugate (ADC), a panel of human tissue factor-specific antibodies (TF HuMab) was generated. Three tissue factor HuMab, that induced efficient inhibition of TF:FVIIa-dependent intracellular signaling, antibody-dependent cell-mediated cytotoxicity, and rapid target internalization, but had minimal impact on tissue factor procoagulant activity in vitro, were conjugated with the cytotoxic agents monomethyl auristatin E (MMAE) or monomethyl auristatin F (MMAF). Tissue factor-specific ADCs showed potent cytotoxicity in vitro and in vivo, which was dependent on tissue factor expression. TF-011-MMAE (HuMax-TF-ADC) was the most potent ADC, and the dominant mechanism of action in vivo was auristatin-mediated tumor cell killing. Importantly, TF-011-MMAE showed excellent antitumor activity in patient-derived xenograft (PDX) models with variable levels of tissue factor expression, derived from seven different solid cancers. Complete tumor regression was observed in all PDX models, including models that showed tissue factor expression in only 25% to 50% of the tumor cells. In conclusion, TF-011-MMAE is a promising novel antitumor agent with potent activity in xenograft models that represent the heterogeneity of human tumors, including heterogeneous target expression.


Subject(s)
Aminobenzoates/therapeutic use , Antineoplastic Agents/therapeutic use , Immunoconjugates/therapeutic use , Neoplasms/drug therapy , Oligopeptides/therapeutic use , Thromboplastin/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Cells, Cultured , HCT116 Cells , HEK293 Cells , Humans , Mice , Mice, Nude , Molecular Targeted Therapy , Thromboplastin/antagonists & inhibitors , Xenograft Model Antitumor Assays
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL