Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Angiogenesis ; 19(3): 389-406, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27234973

RESUMO

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.


Assuntos
Bevacizumab/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/metabolismo , Inibidores da Angiogênese/efeitos adversos , Inibidores da Angiogênese/metabolismo , Inibidores da Angiogênese/uso terapêutico , Animais , Complexo Antígeno-Anticorpo/química , Complexo Antígeno-Anticorpo/metabolismo , Bevacizumab/efeitos adversos , Bevacizumab/uso terapêutico , Linhagem Celular , Células Endoteliais da Veia Umbilical Humana , Humanos , Técnicas In Vitro , Degeneração Macular/imunologia , Degeneração Macular/metabolismo , Degeneração Macular/terapia , Camundongos , Camundongos Transgênicos , Ativação Plaquetária , Ligação Proteica , Multimerização Proteica , Receptores de IgG/genética , Receptores de IgG/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/uso terapêutico , Proteínas Recombinantes de Fusão/efeitos adversos , Proteínas Recombinantes de Fusão/uso terapêutico , Trombocitopenia/etiologia , Trombose/etiologia , Fator A de Crescimento do Endotélio Vascular/imunologia
3.
Thromb Res ; 141: 39-48, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26967531

RESUMO

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.


Assuntos
Micropartículas Derivadas de Células/patologia , Neoplasias Ovarianas/complicações , Neoplasias Ovarianas/diagnóstico , Tromboplastina/análise , Tromboembolia Venosa/complicações , Tromboembolia Venosa/diagnóstico , Adulto , Idoso , Biomarcadores Tumorais/análise , Biomarcadores Tumorais/sangue , Antígeno Ca-125/análise , Antígeno Ca-125/sangue , Feminino , Produtos de Degradação da Fibrina e do Fibrinogênio/análise , Hemostasia , Humanos , Pessoa de Meia-Idade , Neoplasias Ovarianas/sangue , Neoplasias Ovarianas/patologia , Ovário/patologia , Selectina-P/sangue , Período Perioperatório , Período Pré-Operatório , Tromboembolia Venosa/sangue , Tromboembolia Venosa/patologia
4.
Angiogenesis ; 19(1): 79-94, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26650228

RESUMO

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.


Assuntos
Lipocalinas/farmacologia , Terapia de Alvo Molecular , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Complexo Antígeno-Anticorpo/metabolismo , Bevacizumab/farmacologia , Bevacizumab/uso terapêutico , Barreira Hematorretiniana/patologia , Permeabilidade Capilar , Proliferação de Células/efeitos dos fármacos , Feminino , Meia-Vida , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Lipocalinas/farmacocinética , Lipocalinas/uso terapêutico , Camundongos Nus , Camundongos Transgênicos , Mitógenos/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Polietilenoglicóis/química , Engenharia de Proteínas , Coelhos , Receptores de IgG/metabolismo , Transdução de Sinais , Ressonância de Plasmônio de Superfície , Trombose/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
5.
Blood ; 126(19): 2230-8, 2015 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-26396093

RESUMO

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.


Assuntos
Anafilaxia/induzido quimicamente , Anticorpos Monoclonais Murinos/efeitos adversos , Anticorpos Neutralizantes/efeitos adversos , Receptores de IgG/antagonistas & inibidores , Trombocitopenia/induzido quimicamente , Anafilaxia/imunologia , Anafilaxia/patologia , Animais , Anticorpos Monoclonais Murinos/imunologia , Anticorpos Monoclonais Murinos/farmacologia , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/farmacologia , Humanos , Camundongos , Camundongos Transgênicos , Receptores de IgG/genética , Receptores de IgG/imunologia , Trombocitopenia/genética , Trombocitopenia/imunologia , Trombocitopenia/patologia
6.
Arthritis Res Ther ; 17: 234, 2015 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-26335795

RESUMO

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.


Assuntos
Anticorpos Monoclonais/imunologia , Ligante de CD40/imunologia , Imunidade Humoral/imunologia , Trombose/imunologia , Animais , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/química , Anticorpos Monoclonais/farmacologia , Formação de Anticorpos/imunologia , Doenças Autoimunes/imunologia , Doenças Autoimunes/prevenção & controle , Modelos Animais de Doenças , Humanos , Imunidade Humoral/efeitos dos fármacos , Fragmentos Fab das Imunoglobulinas/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/prevenção & controle , Macaca fascicularis , Macaca mulatta , Camundongos Endogâmicos NZB , Polietilenoglicóis/química , Toxoide Tetânico/imunologia , Trombose/induzido quimicamente
7.
Exp Hematol Oncol ; 4: 22, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26251762

RESUMO

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.
J Cardiovasc Nurs ; 29(2): 130-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-23364574

RESUMO

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.


Assuntos
Doenças Cardiovasculares/epidemiologia , Doenças Cardiovasculares/prevenção & controle , Conhecimentos, Atitudes e Prática em Saúde , Lúpus Eritematoso Sistêmico/epidemiologia , Comportamento de Redução do Risco , Idoso , Comunicação , Comorbidade , Feminino , Humanos , Educação de Pacientes como Assunto , Relações Médico-Paciente , Medição de Risco , Fatores de Risco
9.
Cancer Res ; 74(4): 1214-26, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24371232

RESUMO

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.


Assuntos
Aminobenzoatos/uso terapêutico , Antineoplásicos/uso terapêutico , Imunoconjugados/uso terapêutico , Neoplasias/tratamento farmacológico , Oligopeptídeos/uso terapêutico , Tromboplastina/imunologia , Animais , Anticorpos Monoclonais/uso terapêutico , Células Cultivadas , Células HCT116 , Células HEK293 , Humanos , Camundongos , Camundongos Nus , Terapia de Alvo Molecular , Tromboplastina/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Blood Coagul Fibrinolysis ; 24(5): 510-7, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23406663

RESUMO

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.


Assuntos
Coagulação Sanguínea/fisiologia , Neoplasias Ovarianas/diagnóstico , Neoplasias Ovarianas/fisiopatologia , Adulto , Idoso , Biomarcadores Tumorais/sangue , Antígeno Ca-125/sangue , Diagnóstico Diferencial , Feminino , Produtos de Degradação da Fibrina e do Fibrinogênio , Fibrinogênio/análise , Hemostasia , Humanos , Pessoa de Meia-Idade , Período Pré-Operatório , Sensibilidade e Especificidade , Tromboelastografia , Tromboplastina/análise
11.
Blood ; 119(13): 3164-75, 2012 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-22327225

RESUMO

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.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Neoplasias/patologia , Nicho de Células-Tronco/fisiologia , Tromboplastina/farmacologia , Animais , Coagulação Sanguínea/fisiologia , Movimento Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Humanos , Macrófagos/metabolismo , Macrófagos/fisiologia , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Camundongos Transgênicos , Monócitos/metabolismo , Monócitos/fisiologia , Metástase Neoplásica , Neoplasias/metabolismo , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/fisiologia , Nicho de Células-Tronco/efeitos dos fármacos , Tromboplastina/metabolismo
14.
Platelets ; 21(7): 571-4, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20849210

RESUMO

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.


Assuntos
Plaquetas/metabolismo , Receptores do Fator de Necrose Tumoral/sangue , Citometria de Fluxo , Humanos , Receptor de TWEAK
15.
Methods Mol Biol ; 663: 241-59, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20617422

RESUMO

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.


Assuntos
Anticoagulantes/farmacologia , Antineoplásicos/farmacologia , Neoplasias Pulmonares/patologia , Metástase Neoplásica/tratamento farmacológico , Inibidores da Agregação Plaquetária/farmacologia , Animais , Anticoagulantes/uso terapêutico , Antineoplásicos/uso terapêutico , Coagulação Sanguínea/efeitos dos fármacos , Linhagem Celular Tumoral , Enoxaparina/farmacologia , Enoxaparina/uso terapêutico , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Camundongos , Neoplasias Experimentais , Inibidores da Agregação Plaquetária/uso terapêutico , Tromboplastina/antagonistas & inibidores , Varfarina/farmacologia , Varfarina/uso terapêutico
16.
J Immunol ; 185(3): 1577-83, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20585032

RESUMO

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.


Assuntos
Complexo Antígeno-Anticorpo/fisiologia , Autoanticorpos/toxicidade , Ligante de CD40/imunologia , Ativação Plaquetária/imunologia , Receptores de IgG/genética , Trombose/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/toxicidade , Complexo Antígeno-Anticorpo/administração & dosagem , Complexo Antígeno-Anticorpo/toxicidade , Autoanticorpos/administração & dosagem , Autoanticorpos/uso terapêutico , Humanos , Hibridomas , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Ativação Plaquetária/genética , Receptores de IgG/deficiência , Receptores de IgG/fisiologia , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/fisiologia , Proteínas Recombinantes de Fusão/toxicidade , Trombose/sangue
17.
Clin Exp Metastasis ; 26(7): 829-37, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19642003

RESUMO

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.


Assuntos
Antígenos CD40/imunologia , Ligante de CD40/imunologia , Neoplasias Pulmonares/secundário , Melanoma Experimental/patologia , Animais , Transplante de Medula Óssea , Linhagem Celular Tumoral , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Melanoma Experimental/imunologia , Camundongos , Camundongos Knockout , Metástase Neoplásica
18.
Thromb Haemost ; 101(6): 1147-55, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19492160

RESUMO

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.


Assuntos
Reação de Fase Aguda/fisiopatologia , Micropartículas Derivadas de Células/metabolismo , Neoplasias da Próstata/fisiopatologia , Trombina/metabolismo , Tromboplastina/metabolismo , Reação de Fase Aguda/diagnóstico , Reação de Fase Aguda/etiologia , Reação de Fase Aguda/patologia , Idoso , Anticorpos Monoclonais , Coagulação Sanguínea , Testes de Coagulação Sanguínea , Linhagem Celular Tumoral , Separação Celular , Progressão da Doença , Citometria de Fluxo , Humanos , Lipopolissacarídeos/metabolismo , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Estudos Prospectivos , Neoplasias da Próstata/complicações , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/patologia , Sensibilidade e Especificidade , Trombina/genética , Tromboplastina/imunologia
19.
Semin Thromb Hemost ; 33(7): 643-52, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18000790

RESUMO

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.


Assuntos
Hemostasia , Lipoproteínas/metabolismo , Lipoproteínas/farmacologia , Metástase Neoplásica , Neoplasias/sangue , Neoplasias/patologia , Tromboplastina/antagonistas & inibidores , Animais , Anticoagulantes/metabolismo , Anticoagulantes/farmacologia , Fibrinolíticos/metabolismo , Fibrinolíticos/farmacologia , Heparina de Baixo Peso Molecular/metabolismo , Heparina de Baixo Peso Molecular/farmacologia , Humanos , Metástase Neoplásica/prevenção & controle , Neoplasias/irrigação sanguínea , Neovascularização Patológica , Inibidores de Serina Proteinase/metabolismo , Inibidores de Serina Proteinase/farmacologia , Tromboplastina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...