Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 678
Filtrar
1.
Oxid Med Cell Longev ; 2021: 3827501, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34804364

RESUMO

Acute lung injury (ALI) is one of the fatal symptoms of sepsis. However, there were no effective clinical treatments. TF accumulation-induced fibrin deposit formations and coagulation abnormalities in pulmonary vessels contribute to the lethality of ALI. Suppressor of cytokine signaling 3 (SOCS3) acts as an endogenous negative regulator of the TLR4/TF pathway. We hypothesized that inducing SOCS3 expression using lidocaine to suppress the TLR4/TF pathway may alleviate ALI. Hematoxylin and eosin (H&E), B-mode ultrasound, and flow cytometry were used to measure the pathological damage of mice. Gelatin zymography was used to measure matrix metalloproteinase-2/9 (MMP-2/9) activities. Western blot was used to assay the expression of protein levels. Here, we show that lidocaine could increase the survival rate of ALI mice and ameliorate the lung injury of ALI mice including reducing the edema, neutrophil infiltration, and pulmonary thrombosis formation and increasing blood flow velocity. Moreover, in vitro and in vivo, lidocaine could increase the expression of p-AMPK and SOCS3 and subsequently decrease the expression of p-ASK1, p-p38, TF, and the activity of MMP-2/9. Taken together, our study demonstrated that lidocaine could inhibit the TLR4/ASK1/TF pathway to alleviate ALI via activating AMPK-SOCS3 axis.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Regulação da Expressão Gênica/efeitos dos fármacos , Lidocaína/farmacologia , Metaloproteinase 2 da Matriz/química , Metaloproteinase 9 da Matriz/química , Sepse/complicações , Tromboplastina/antagonistas & inibidores , Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/patologia , Animais , Lipopolissacarídeos/toxicidade , Masculino , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Tromboplastina/genética , Tromboplastina/metabolismo , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia
2.
Int J Mol Sci ; 22(21)2021 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-34769515

RESUMO

Cancer-associated thrombosis is the second-leading cause of mortality in patients with cancer and presents a poor prognosis, with a lack of effective treatment strategies. NAD(P)H quinone oxidoreductase 1 (NQO1) increases the cellular nicotinamide adenine dinucleotide (NAD+) levels by accelerating the oxidation of NADH to NAD+, thus playing important roles in cellular homeostasis, energy metabolism, and inflammatory responses. Using a murine orthotopic 4T1 breast cancer model, in which multiple thrombi are generated in the lungs at the late stage of cancer development, we investigated the effects of regulating the cellular NAD+ levels on cancer-associated thrombosis. In this study, we show that dunnione (a strong substrate of NQO1) attenuates the prothrombotic state and lung thrombosis in tumor-bearing mice by inhibiting the expression of tissue factor and formation of neutrophil extracellular traps (NETs). Dunnione increases the cellular NAD+ levels in lung tissues of tumor-bearing mice to restore the declining sirtuin 1 (SIRT1) activity, thus deacetylating nuclear factor-kappa B (NF-κB) and preventing the overexpression of tissue factor in bronchial epithelial and vascular endothelial cells. In addition, we demonstrated that dunnione abolishes the ability of neutrophils to generate NETs by suppressing histone acetylation and NADPH oxidase (NOX) activity. Overall, our results reveal that the regulation of cellular NAD+ levels by pharmacological agents may inhibit pulmonary embolism in tumor-bearing mice, which may potentially be used as a viable therapeutic approach for the treatment of cancer-associated thrombosis.


Assuntos
Neoplasias da Mama/complicações , Armadilhas Extracelulares/efeitos dos fármacos , NAD(P)H Desidrogenase (Quinona)/metabolismo , NAD/metabolismo , Naftoquinonas/farmacologia , Trombofilia/tratamento farmacológico , Tromboplastina/metabolismo , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Sirtuína 1/metabolismo , Trombofilia/etiologia , Trombofilia/prevenção & controle , Tromboplastina/antagonistas & inibidores , Trombose/tratamento farmacológico , Trombose/etiologia , Trombose/prevenção & controle
3.
Tohoku J Exp Med ; 255(1): 1-8, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34511578

RESUMO

Endothelial nitric oxide synthase (eNOS) dysfunction is known to exacerbate the progression and prognosis of diabetic kidney disease (DKD). One of the mechanisms through which this is achieved is that low eNOS levels are associated with hypercoagulability, which promotes kidney injury. In the extrinsic coagulation cascade, the tissue factor (factor III) and downstream coagulation factors, such as active factor X (FXa), exacerbate inflammation through activation of the protease-activated receptors (PARs). Recently, it has been shown that the lack of or reduced eNOS expression in diabetic mice, as a model of advanced DKD, increases renal tissue factor levels and PAR1 and 2 expression in their kidneys. Furthermore, pharmaceutical inhibition or genetic deletion of coagulation factors or PARs ameliorated inflammation in DKD in mice lacking eNOS. In this review, we summarize the relationship between eNOS, coagulation, and PARs and propose a novel therapeutic option for the management of patients with DKD.


Assuntos
Nefropatias Diabéticas/etiologia , Óxido Nítrico Sintase Tipo III/deficiência , Receptores Ativados por Proteinase/metabolismo , Animais , Anticorpos Neutralizantes/administração & dosagem , Coagulação Sanguínea , Nefropatias Diabéticas/sangue , Nefropatias Diabéticas/fisiopatologia , Modelos Animais de Doenças , Inibidores do Fator Xa/farmacologia , Humanos , Rim/efeitos dos fármacos , Rim/metabolismo , Camundongos , Camundongos Knockout , Óxido Nítrico Sintase Tipo III/genética , Receptores Ativados por Proteinase/deficiência , Receptores Ativados por Proteinase/genética , Transdução de Sinais , Tromboplastina/antagonistas & inibidores , Tromboplastina/metabolismo
4.
Biochem Pharmacol ; 192: 114671, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34246626

RESUMO

Sepsis-induced disseminated intravascular coagulation (DIC) is a common life-threatening terminal-stage disease with high mortality. This study aimed to identify effective miRNAs as therapeutic targets for DIC. Bioinformatics and luciferase reporter gene analyses were performed to predict miR-19a-3p and validate that it targets tissue factor (TF). Quantitative real-time PCR was used to detect the expression of miR-19a-3p and TF, and TF procoagulant activity was determined using the chromogenic substrate method. Western blotting was used to detect the protein levels of TF, AKT serine/threonine kinase (AKT), extracellular regulated protein kinases (ERK), nuclear factor kappa B (NF-κB) P65, NFKB inhibitor alpha (IκB-a) and their phosphorylated counterparts in cell experiments. Furthermore, a rat model was established to explore the potential of miR-19a-3p in DIC treatment. As a result, a human clinical study revealed that miR-19a-3p was downregulated and that TF was upregulated in neonates with sepsis-induced DIC compared with those in the control group. The luciferase reporter assay showed that TF was a direct target of miR-19a-3p. Cell experiments verified that the mRNA and protein levels of TF, and the p-AKT/AKT, p-Erk/Erk, p-P65/P65, p-IκB-a/IκB-a ratios, and TF procoagulant activity were significantly decreased in lipopolysaccharide (LPS) -induced human peripheral blood mononuclear cells (PBMCs) and human umbilical vein endothelial cells (HUVECs) inhibited by overexpression of miR-19a-3p, and that miR-19a-3p regulating TF was dependent on the NF-kB and AKT pathways. In vivo, miR-19a-3p injection into DIC rats suppressed the mRNA expression of TF; more importantly, significant improvements in coagulation function indicators and in histopathologies of lung and kidney were observed. In conclusion, miR-19a-3p may suppress DIC by targeting TF and might be a potential therapeutic target in treating sepsis-induced DIC.


Assuntos
Coagulação Intravascular Disseminada/metabolismo , Regulação para Baixo/fisiologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , MicroRNAs/biossíntese , Sepse/metabolismo , Tromboplastina/metabolismo , Animais , Células Cultivadas , Coagulação Intravascular Disseminada/induzido quimicamente , Regulação para Baixo/efeitos dos fármacos , Feminino , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Recém-Nascido , Lipopolissacarídeos/toxicidade , Masculino , Ratos , Ratos Sprague-Dawley , Sepse/induzido quimicamente , Tromboplastina/antagonistas & inibidores
5.
Oncol Rep ; 45(1): 329-336, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33200231

RESUMO

Tissue factor (TF) is an attractive target for cancer therapy due to its overexpression in multiple types of malignancies. In addition, TF has been reported to play functional roles in both cancer development and metastasis. Several groups have already developed antibody­drug conjugates (ADCs) against TF for use as cancer treatments, and have demonstrated their efficacies in conventional subcutaneous xenograft models and patient­derived xenograft models. However, no previous studies have investigated the effectiveness of anti­TF ADC in an advanced­stage cancer model. The present study developed an original humanized anti­TF monoclonal antibody conjugated with monomethyl auristatin E, and evaluated its in vivo efficacy in a pancreatic cancer xenograft model with peritoneal dissemination. In vitro assays demonstrated that the anti­TF ADC had potent binding affinity and cytotoxic activity against human pancreatic cancer cells that strongly expressed TF antigens. The anti­TF ADC also exhibited greater antitumor effect than that of a control ADC in conventional subcutaneous xenograft models, with efficacy depending on the TF expression in the tumor tissues. Furthermore, the anti­TF ADC significantly inhibited tumor growth in an orthotopic xenograft model, and extended the survival period in a murine peritoneal dissemination model. These results indicated that anti­TF ADC has the potential to be an effective treatment not only for primary tumors, but also for those that are widely disseminated. Therefore, it can be concluded that ADC targeting TF may be a promising agent for advanced pancreatic cancer therapy.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Imunoconjugados/uso terapêutico , Neoplasias Pancreáticas/tratamento farmacológico , Tromboplastina/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Pancreáticas/mortalidade , Neoplasias Pancreáticas/patologia , Peritônio/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
6.
J Hematol Oncol ; 13(1): 93, 2020 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-32665005

RESUMO

Tissue factor (TF) is the primary initiator of the coagulation cascade, though its effects extend well beyond hemostasis. When TF binds to Factor VII, the resulting TF:FVIIa complex can proteolytically cleave transmembrane G protein-coupled protease-activated receptors (PARs). In addition to activating PARs, TF:FVIIa complex can also activate receptor tyrosine kinases (RTKs) and integrins. These signaling pathways are utilized by tumors to increase cell proliferation, angiogenesis, metastasis, and cancer stem-like cell maintenance. Herein, we review in detail the regulation of TF expression, mechanisms of TF signaling, their pathological consequences, and how it is being targeted in experimental cancer therapeutics.


Assuntos
Proteínas de Neoplasias/fisiologia , Neoplasias/sangue , Trombofilia/sangue , Tromboplastina/fisiologia , Sequência de Aminoácidos , Hipóxia Celular , Fator VIIa/fisiologia , Regulação Neoplásica da Expressão Gênica , Humanos , Imunoterapia Adotiva , Integrinas/metabolismo , Dados de Sequência Molecular , Terapia de Alvo Molecular , Invasividade Neoplásica , Metástase Neoplásica , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias/irrigação sanguínea , Neoplasias/fisiopatologia , Neoplasias/terapia , Células-Tronco Neoplásicas/patologia , Neovascularização Patológica/fisiopatologia , Conformação Proteica , Domínios Proteicos , Isoformas de Proteínas/fisiologia , Receptores Proteína Tirosina Quinases/metabolismo , Receptores Ativados por Proteinase/metabolismo , Transdução de Sinais/fisiologia , Trombofilia/etiologia , Tromboplastina/antagonistas & inibidores
7.
Naunyn Schmiedebergs Arch Pharmacol ; 393(10): 1887-1898, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32430618

RESUMO

BACKGROUND: Retinoid receptors (RRs), RAR-α and RXR-α, work as transcription factors that regulate cell growth, differentiation, survival, and death. Hepatic stellate cells (HSCs) store retinoid and release its RRs as lipid droplets upon their activation. PURPOSE: We test the hypothesis that loss of retinoid receptors RAR-α and RXR-α from HSCs is dependent on tissue factor (TF) during thioacetamide (TAA)-induced liver injury. METHODS: Liver toxicity markers, TF, fibrin, cleaved caspase-3, and cyclin D1 as well as histopathology were investigated. RESULTS: Increased TF, fibrin, cleaved caspase-3, and cyclin D1 protein expression is seen in zone of central vein after TAA injection compared with vehicle-treated mice. A strong downregulation of RAR-α and RXR-α is seen in TAA-induced liver injury. In addition, histopathological obliteration and pericentral expression of cleaved caspase 3 and cyclin D1 are observed after TAA injection compared with the normal vehicle-treated mice. No changes have been seen in TAA/TF-sense (SC) in whole parameters compared with TAA-treated animals. TAA/TF-antisense (AS)-treated mice show normal expression of all parameters and normal histopathological features when compared with the control mice. In conclusion, this study declares that the strong downregulation of RAR-α and RXR-α may cause liver injury and particularly activation of HSCs in TAA-induced toxicity. TF-AS treatment not only downregulates TF protein expression but also alleviates loss of liver RAR-α and RXR-α and suppresses the activated apoptosis signals in TAA-induced liver toxicity. Finally, TF and RAR-α/RXR-α are important regulatory molecules in TAA induced acute liver injury.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Tioacetamida/toxicidade , Tromboplastina/antagonistas & inibidores , Tromboplastina/metabolismo , Animais , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Masculino , Camundongos , Membro 1 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Receptor X Retinoide alfa/metabolismo
8.
Adv Exp Med Biol ; 1221: 771-784, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32274737

RESUMO

The hemostatic cascade is initiated by the transmembrane coagulation protein - tissue factor (TF) and eventuates in fibrin formation. Heparanase protein was demonstrated to directly enhance TF activity resulting in increased activation of the coagulation system. In addition, heparanase was found to increase hemostatic system activation via two other mechanisms: up-regulating TF expression in endothelial cells and releasing the protein tissue factor pathway inhibitor (TFPI) from the cell surface. Peptides derived from TFPI-2, a protein similar to TFPI, were shown to inhibit the TF/heparanase complex as well as attenuate sepsis and tumor growth. Increased heparanase procoagulant activity was observed in several clinical settings, including women using oral contraceptives, women at delivery, patients following orthopedic surgery and patients with diabetic foot, shift work female nurses, patients with lung cancer, retinal vein thrombosis and prosthetic heart valve thrombosis. Remarkably, the heparanase profile was significantly different across the tested groups. Inhibition of TF / heparanase interaction may represent a new target for attenuating coagulation, cancer and inflammation.


Assuntos
Coagulação Sanguínea , Glucuronidase/metabolismo , Células Endoteliais/metabolismo , Glucuronidase/antagonistas & inibidores , Humanos , Inflamação , Neoplasias , Tromboplastina/antagonistas & inibidores , Tromboplastina/metabolismo
9.
Mol Pharm ; 17(5): 1697-1705, 2020 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-32202792

RESUMO

Pancreatic cancer is highly aggressive, with a median survival time of less than 6 months and a 5-year overall survival rate of around 7%. The poor prognosis of PaCa is largely due to its advanced stage at diagnosis and the lack of efficient therapeutic options. Thus, the development of an efficient, multifunctional PaCa theranostic system is urgently needed. Overexpression of tissue factor (TF) has been associated with increased tumor growth, angiogenesis, and metastasis in many malignancies, including pancreatic cancer. Herein, we propose the use of a TF-targeted monoclonal antibody (ALT836) conjugated with the pair 86/90Y as a theranostic agent against pancreatic cancer. For methods, serial PET imaging with 86Y-DTPA-ALT836 was conducted to map the biodistribution the tracer in BXPC-3 tumor-bearing mice. 90Y-DTPA-ALT836 was employed as a therapeutic agent that also allowed tumor burden monitoring through Cherenkov luminescence imaging. The results were that the uptake of 86Y-DTPA-ALT836 in BXPC-3 xenograft tumors was high and increased over time up to 48 h postinjection (p.i.), corroborated through ex vivo biodistribution studies and further confirmed by Cherenkov luminescence Imaging. In therapeutic studies, 90Y-DTPA-ALT836 was found to slow tumor growth relative to the control groups and had significantly smaller (p < 0.05) tumor volumes 1 day p.i. Histological analysis of ex vivo tissues revealed significant damage to the treated tumors. The conclusion is that the use of the 86/90Y theranostic pair allows PET imaging with excellent tumor-to-background contrast and treatment of TF-expressing pancreatic tumors with promising therapeutic outcomes.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Neoplasias Pancreáticas/tratamento farmacológico , Tromboplastina/antagonistas & inibidores , Radioisótopos de Ítrio/farmacocinética , Animais , Anticorpos Monoclonais/farmacocinética , Linhagem Celular Tumoral , Feminino , Camundongos , Neoplasias Pancreáticas/patologia , Tomografia por Emissão de Pósitrons , Distribuição Tecidual
10.
Toxins (Basel) ; 11(7)2019 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-31331004

RESUMO

The genus Bitis comprises 17 snake species that inhabit Africa and the Arabian Peninsula. They are responsible for a significant proportion of snakebites in the region. The venoms of the two independent lineages of giant Bitis (B. arietans and again in the common ancestor of the clade consisting of B. gabonica, B. nasicornis, B. parviocula and B. rhinoceros) induce an array of debilitating effects including anticoagulation, hemorrhagic shock and cytotoxicity, whilst the dwarf species B. atropos is known to have strong neurotoxic effects. However, the venom effects of the other species within the genus have not been explored in detail. A series of coagulation assays were implemented to assess the coagulotoxic venom effects of fourteen species within the genus. This study identified procoagulant venom as the ancestral condition, retained only by the basal dwarf species B. worthingtoni, suggesting anticoagulant venom is a derived trait within the Bitis genus and has been secondarily amplified on at least four occasions. A wide range of anticoagulant mechanisms were identified, such as coagulant and destructive activities upon fibrinogen in both giant and dwarf Bitis and the action of inhibiting the prothrombinase complex, which is present in a clade of dwarf Bitis. Antivenom studies revealed that while the procoagulant effects of B. worthingtoni were poorly neutralized, and thus a cause for concern, the differential mechanisms of anticoagulation in other species were all well neutralized. Thus, this study concludes there is a wide range of coagulotoxic mechanisms which have evolved within the Bitis genus and that clinical management strategies are limited for the procoagulant effects of B. worthingtoni, but that anticoagulant effects of other species are readily treated by the South African polyvalent antivenom. These results therefore have direct, real-work implications for the treatment of envenomed patients.


Assuntos
Anticoagulantes/toxicidade , Antivenenos/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Coagulantes/toxicidade , Venenos de Víboras/toxicidade , Viperidae , Animais , Fibrinogênio/metabolismo , Humanos , Tromboelastografia , Tromboplastina/antagonistas & inibidores
11.
Haemophilia ; 25(5): 797-806, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31336410

RESUMO

INTRODUCTION: Tissue factor pathway inhibitor (TFPI) is an endogenous inhibitor of the extrinsic pathway that negatively regulates thrombin production during coagulation. Under haemophilic conditions, where the intrinsic coagulation pathway is impaired, inhibition of TFPI may improve clotting. AIM: We investigated the ex vivo effects of a human TFPI neutralizing antibody, marstacimab (previously PF-06741086), in coagulation assays including rotational thromboelastometry (ROTEM), thrombin generation assay (TGA) and the dilute prothrombin time (dPT) assay, performed in haemophilic whole blood and plasmas. We compared the effects of marstacimab to the effects of recombinant coagulation factors and investigated the reproducibility of marstacimab in restoring haemostasis by comparing its effect in whole blood collected from the same study participants on differing days. METHODS: Citrated whole blood and plasmas obtained from haemophilia participants were supplemented ex vivo with vehicle, marstacimab, recombinant FVIII (rFVIII) or recombinant factor IX (rFIX) and analysed in ROTEM, TGA and the dPT assay using low tissue factor concentrations to trigger coagulation. RESULTS: Marstacimab induced pro-coagulant responses in ROTEM parameters including reduction in clotting times and increases in angle. Similarly, participant plasmas supplemented with marstacimab exhibited improvements in TGA parameters, including reduced lag times, increased peak thrombin concentrations and reductions in dPT clotting time. Concentrations of marstacimab tested showed activity comparable to addition of rFVIII or rFIX and were reproducible. CONCLUSIONS: These studies show the ex vivo potency of marstacimab in restoring haemostasis in whole blood and plasmas from haemophilia participants and comparability to ex vivo reconstitution with recombination coagulation factors.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Coagulação Sanguínea/efeitos dos fármacos , Hemofilia A/tratamento farmacológico , Plasma/metabolismo , Tromboplastina/antagonistas & inibidores , Anticorpos Monoclonais Humanizados/farmacologia , Feminino , Hemofilia A/patologia , Humanos , Masculino
12.
Cancer Sci ; 110(10): 3296-3305, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31348600

RESUMO

Tissue factor (TF) is known to be overexpressed in various cancers including pancreatic cancer. The upregulation of TF expression has been observed not only in tumor cells, but also in tumor stromal cells. Because of the potential of TF as a delivery target, several studies investigated the effectiveness of Ab-drug conjugates (ADCs) against TF for cancer therapy. However, it is still unclear whether anti-TF ADC can exert toxicity against both tumor cells and tumor stromal cells. Here, we prepared ADC using a rat anti-mouse TF mAb (clone.1157) and 2 types of in vivo murine pancreatic cancer models, one s.c. and other orthotopic with an abundant tumor stroma. We also compared the feasibility of bis-alkylating conjugation (bisAlk) with that of conventional maleimide-based conjugation (MC). In the s.c. models, anti-TF ADC showed greater antitumor effects than control ADC. The results also indicated that the bisAlk linker might be more suitable than the MC linker for cancer treatments. In the orthotopic model, anti-TF ADC showed greater in vivo efficacy and more extended survival time control ADC. Treatment with anti-TF ADC (20 mg/kg, three times a week) did not affect mouse body weight changes in any in vivo experiment. Furthermore, immunofluorescence staining indicated that anti-TF ADC delivered agents not only to TF-positive tumor cells, but also to TF-positive tumor vascular endothelial cells and other tumor stromal cells. We conclude that anti-TF ADC should be a selective and potent drug for pancreatic cancer therapy.


Assuntos
Alquilantes/química , Antineoplásicos Imunológicos/administração & dosagem , Imunoconjugados/administração & dosagem , Maleimidas/química , Neoplasias Pancreáticas/tratamento farmacológico , Tromboplastina/antagonistas & inibidores , Animais , Antineoplásicos Imunológicos/química , Antineoplásicos Imunológicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Esquema de Medicação , Feminino , Humanos , Imunoconjugados/química , Imunoconjugados/farmacologia , Camundongos , Camundongos Transgênicos , Neoplasias Pancreáticas/metabolismo , Ratos , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Células Estromais/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Semin Thromb Hemost ; 45(4): 385-395, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31096306

RESUMO

There is a strong relationship between tissue factor (TF) and cancer. Many cancer cells express high levels of both full-length TF and alternatively spliced (as) TF. TF expression in cancer is associated with poor prognosis. In this review, the authors summarize the regulation of TF expression in cancer cells and the roles of TF and asTF in tumor growth and metastasis. A variety of different signaling pathways, transcription factors and micro ribonucleic acids regulate TF gene expression in cancer cells. The TF/factor VIIa complex enhances tumor growth by activating protease-activated receptor 2 signaling and by increasing the expression of angiogenic factors, such as vascular endothelial growth factor. AsTF increases tumor growth by enhancing integrin ß1 signaling. TF and asTF also contribute to metastasis via multiple thrombin-dependent and independent mechanisms that include protecting tumor cells from natural killer cells. Finally, a novel anticancer therapy is using tumor TF as a target to deliver cytotoxic drugs to the tumor. TF may be useful in diagnosis, prognosis, and treatment of cancer.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias/genética , Transdução de Sinais/genética , Tromboplastina/genética , Humanos , Imunoconjugados/uso terapêutico , Terapia de Alvo Molecular/métodos , Metástase Neoplásica , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Prognóstico , Transdução de Sinais/efeitos dos fármacos , Tromboplastina/antagonistas & inibidores , Tromboplastina/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
14.
Gynecol Oncol ; 153(3): 694-702, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30929824

RESUMO

Antibody drug conjugates (ADCs) are an exciting class of oncologic therapeutics. ADCs have been FDA approved in hematologic malignancies and breast cancer and are a growing area of study in numerous solid malignancies. The desire for tumor-specific therapies with decreased systemic toxicity has driven over a decade of research into the design and optimization of ADCs, which are now in a third generation of development. Gynecologic malignancies in particular suffer a dearth of novel therapies. This review will examine the field of ADCs in gynecologic cancers, focusing on ADCs targeting folate receptor alpha (FRα), mesothelin, tissue factor, MUC16 (CA125), NaPi2B, and Trop2.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Neoplasias dos Genitais Femininos/tratamento farmacológico , Imunoconjugados/uso terapêutico , Maitansina/análogos & derivados , Antígenos de Neoplasias , Antígeno Ca-125 , Camptotecina/análogos & derivados , Camptotecina/uso terapêutico , Moléculas de Adesão Celular/antagonistas & inibidores , Desenho de Fármacos , Feminino , Receptor 1 de Folato/antagonistas & inibidores , Proteínas Ligadas por GPI/antagonistas & inibidores , Humanos , Maitansina/uso terapêutico , Proteínas de Membrana/antagonistas & inibidores , Mesotelina , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/antagonistas & inibidores , Tromboplastina/antagonistas & inibidores
15.
J Control Release ; 284: 49-56, 2018 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-29906553

RESUMO

Antibody-drug conjugates (ADCs) are currently considered to be promising agents for cancer therapy. However, especially in solid tumors, the uneven distribution of ADCs would decrease their efficacy in clinical studies. We suggest that in addition to optimizing ADC components, such as the linker structure and anticancer agent, it is necessary to consider the distribution of the ADC within tumor tissue. In this study, we established three kinds of anti-tissue factor (TF) ADCs: 1849ADC with a low kd, 444ADC with an intermediate kd, and 1084ADC with a high kd. All three of the anti-TF ADCs exhibited almost the same in vitro cytotoxicity and pharmacological and biochemical characteristics, although the binding kinetics parameters differed. In vivo, all ADCs exerted equivalent antitumor effects against small BxPC3 tumors. However, on larger BxPC3 tumors, 1084ADC (higher kd) exerted higher antitumor activity than 1849ADC (lower kd). Furthermore, immunofluorescence staining indicated that 1084ADC was distributed throughout the whole tumor, whereas 1849ADC was mainly localized close to tumor vessels. We conclude that the ADC with a higher kd increased the antitumor effect of because it penetrated and distributed evenly throughout the entire solid tumor. These findings highlight the importance of the kd of a mAb in ADC design.


Assuntos
Antineoplásicos Imunológicos/farmacocinética , Antineoplásicos Imunológicos/uso terapêutico , Imunoconjugados/farmacocinética , Imunoconjugados/uso terapêutico , Neoplasias Pancreáticas/tratamento farmacológico , Tromboplastina/antagonistas & inibidores , Animais , Antineoplásicos Imunológicos/química , Linhagem Celular Tumoral , Feminino , Humanos , Imunoconjugados/química , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Tromboplastina/metabolismo
16.
Biomacromolecules ; 19(6): 2320-2329, 2018 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-29767505

RESUMO

Antibody fragment (Fab')-installed polyion complex (PIC) micelles were constructed to improve targetability of small interfering RNA (siRNA) delivery to pancreatic cancer cells. To this end, we synthesized a block copolymer of azide-functionalized poly(ethylene glycol) and poly(l-lysine) and prepared PIC micelles with siRNA. Then, a dibenzylcyclooctyne (DBCO)-modified antihuman tissue factor (TF) Fab' was conjugated to azido groups on the micellar surface. A fluorescence correlation spectroscopic analysis revealed that 1, 2, or 3 molecule(s) of Fab'(s) were installed onto one micellar nanoparticle according to the feeding ratio of Fab' (or DBCO) to micelle (or azide). The resulting micelles exhibited ∼40 nm in hydrodynamic diameter, similar to that of the parent micelles before Fab' conjugation. Flow cytometric analysis showed that three molecules of Fab'-installed PIC micelles (3(Fab')-micelles) had the highest binding affinity to cultured pancreatic cancer BxPC3 cells, which are known to overexpress TF on their surface. The 3(Fab')-micelles also exhibited the most efficient gene silencing activity against polo-like kinase 1 mRNA in the cultured cancer cells. Furthermore, the 3(Fab')-micelles exhibited high penetrability and the highest cellular internalization amounts in BxPC3 spheroids compared with one or two molecule(s) of Fab'-installed PIC micelles. These results demonstrate the potential of anti-TF Fab'-installed PIC micelles for active targeting of stroma-rich pancreatic tumors.


Assuntos
Anticorpos Antineoplásicos , Proteínas de Ciclo Celular/antagonistas & inibidores , Sistemas de Liberação de Medicamentos , Inativação Gênica , Fragmentos Fab das Imunoglobulinas , Micelas , Neoplasias Pancreáticas/tratamento farmacológico , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores , RNA Interferente Pequeno , Tromboplastina/antagonistas & inibidores , Anticorpos Antineoplásicos/química , Anticorpos Antineoplásicos/farmacologia , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Humanos , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/farmacologia , Neoplasias Pancreáticas/enzimologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Polietilenoglicóis/química , Polietilenoglicóis/farmacologia , Polilisina/química , Polilisina/farmacologia , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/genética , RNA Interferente Pequeno/química , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Tromboplastina/metabolismo , Quinase 1 Polo-Like
17.
Anticancer Res ; 38(5): 2635-2642, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29715083

RESUMO

BACKGROUND/AIM: Tissue factor (TF) expression increases cancer stem cell (CSC) activity in breast and lung cancer. There are ongoing studies focused on targeting CSCs via anti-TF treatment, for breast and lung cancer therapy. Herein, the aim was to determine whether targeting TF could have an anti-CSC therapeutic role in colorectal cancer (CRC). MATERIALS AND METHODS: Evaluation of colonosphere-forming efficiency (CFE) and aldehyde dehydrogenase (ALDH) expression level was used to quantify CSC activity in two CRC cell lines, after TF knockdown (TFKD) or TF over-expression (TFOE). RESULTS: TFKD resulted in increased levels of ALDH in SW620 (1.31±0.04-fold, p<0.001) and DLD-1 (1.63±0.14-fold, p=0.04) cells. CFE was increased in SW620 (1.21±0.23% vs. 2.03±0.29%, p=0.01) and DLD-1 (0.41±0.12% vs. 0.68±0.9%, p=0.01) cells. Conversely, TFOE decreased ALDH expression (0.72±0.04-fold, p=0.001) and CFE (0.33±0.05% vs. 0.66±0.14%, p=0.006) in DLD-1, but had no impact on SW620 cells. CONCLUSION: In the examined CRC cell lines, TF expression was inversely related to CSC activity suggesting that anti-TF therapies may not have a role in CRC treatment.


Assuntos
Neoplasias Colorretais/patologia , Células-Tronco Neoplásicas/patologia , Tromboplastina/fisiologia , Aldeído Desidrogenase/análise , Biomarcadores Tumorais , Divisão Celular , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Vetores Genéticos/farmacologia , Humanos , Lentivirus/genética , Células-Tronco Neoplásicas/metabolismo , Interferência de RNA , RNA Interferente Pequeno/genética , Proteínas Recombinantes/metabolismo , Esferoides Celulares , Tromboplastina/antagonistas & inibidores , Tromboplastina/genética
18.
Cancer Immunol Res ; 6(6): 671-684, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29622581

RESUMO

Triple-negative breast cancer (TNBC) is a leading cause of breast cancer death and is often associated with BRCA1 and BRCA2 mutation. Due to the lack of validated target molecules, no targeted therapy for TNBC is approved. Tissue factor (TF) is a common yet specific surface target receptor for cancer cells, tumor vascular endothelial cells, and cancer stem cells in several types of solid cancers, including breast cancer. Here, we report evidence supporting the idea that TF is a surface target in TNBC. We used in vitro cancer lines and in vivo tumor xenografts in mice, all with BRCA1 or BRCA2 mutations, derived from patients' tumors. We showed that TF is overexpressed on TNBC cells and tumor neovasculature in 50% to 85% of TNBC patients (n = 161) and in TNBC cell line-derived xenografts (CDX) and patient-derived xenografts (PDX) from mice, but was not detected in adjacent normal breast tissue. We then describe the development of a second-generation TF-targeting immunoconjugate (called L-ICON1, for lighter or light chain ICON) with improved efficacy and safety profiles compared with the original ICON. We showed that L-ICON1 kills TNBC cells in vitro via antibody-dependent cell-mediated cytotoxicity and can be used to treat human and murine TNBC CDX as well as PDX in vivo in orthotopic mouse models. Thus, TF could be a useful target for the development of immunotherapeutics for TNBC patients, with or without BRCA1 and BRCA2 mutations. Cancer Immunol Res; 6(6); 671-84. ©2018 AACR.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Imunoconjugados/farmacologia , Tromboplastina/antagonistas & inibidores , Neoplasias de Mama Triplo Negativas/imunologia , Neoplasias de Mama Triplo Negativas/metabolismo , Animais , Biomarcadores Tumorais , Células CHO , Linhagem Celular Tumoral , Cricetulus , Modelos Animais de Doenças , Feminino , Expressão Gênica , Humanos , Imuno-Histoquímica , Terapia de Alvo Molecular , Mutação , Tromboplastina/genética , Tromboplastina/metabolismo , Neoplasias de Mama Triplo Negativas/diagnóstico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Ensaios Antitumorais Modelo de Xenoenxerto
19.
JCI Insight ; 3(6)2018 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-29563336

RESUMO

Hemostatic defects are treated using coagulation factors; however, clot formation also requires a procoagulant phospholipid (PL) surface. Here, we show that innate immune cell-derived enzymatically oxidized phospholipids (eoxPL) termed hydroxyeicosatetraenoic acid-phospholipids (HETE-PLs) restore hemostasis in human and murine conditions of pathological bleeding. HETE-PLs abolished blood loss in murine hemophilia A and enhanced coagulation in factor VIII- (FVIII-), FIX-, and FX-deficient human plasma . HETE-PLs were decreased in platelets from patients after cardiopulmonary bypass (CPB). To explore molecular mechanisms, the ability of eoxPL to stimulate individual isolated coagulation factor/cofactor complexes was tested in vitro. Extrinsic tenase (FVIIa/tissue factor [TF]), intrinsic tenase (FVIIIa/FIXa), and prothrombinase (FVa/FXa) all were enhanced by both HETE-PEs and HETE-PCs, suggesting a common mechanism involving the fatty acid moiety. In plasma, 9-, 15-, and 12-HETE-PLs were more effective than 5-, 11-, or 8-HETE-PLs, indicating positional isomer specificity. Coagulation was enhanced at lower lipid/factor ratios, consistent with a more concentrated area for protein binding. Surface plasmon resonance confirmed binding of FII and FX to HETE-PEs. HETE-PEs increased membrane curvature and thickness, but not surface charge or homogeneity, possibly suggesting increased accessibility to cations/factors. In summary, innate immune-derived eoxPL enhance calcium-dependent coagulation factor function, and their potential utility in bleeding disorders is proposed.


Assuntos
Fatores de Coagulação Sanguínea/metabolismo , Hemorragia/enzimologia , Hemorragia/metabolismo , Fosfolipídeos/metabolismo , Trombina/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Coagulação Sanguínea , Fatores de Coagulação Sanguínea/genética , Plaquetas , Ponte Cardiopulmonar/efeitos adversos , Proteínas de Transporte , Cisteína Endopeptidases , Fator IX/genética , Fator VIII/genética , Fator VIIa/metabolismo , Fator X/genética , Hemofilia A , Hemorragia/prevenção & controle , Hemostasia , Humanos , Ácidos Hidroxieicosatetraenoicos , Lipoproteínas/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Proteínas de Neoplasias , Ressonância de Plasmônio de Superfície , Tromboplastina/antagonistas & inibidores , Tromboplastina/metabolismo
20.
Reprod Biol ; 18(1): 109-114, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29422377

RESUMO

Endometriosis is a common condition in reproductive-aged women characterized by ectopic endometrial lesions of varied appearance, including red, white, blue, black or powder burn coloration, which contribute to chronic pain and infertility. The immunoconjugate molecule (Icon) targets Tissue Factor, a transmembrane receptor for Factor VII/VIIa that is aberrantly expressed in the endothelium supporting ectopic endometrial tissue. Icon has been shown to cause regression of endometriosis in a murine model of disease but prior to this study had not been tested in non-human primates. This study evaluated Icon as a novel treatment for endometriosis in non-human primates (Papio anubis) using an adenoviral vector (AdIcon) delivery system. Female baboons (n = 15) underwent surgical induction of endometriosis. After laparoscopic confirmation of endometriosis lesions 6-weeks post-surgery, the treatment group (n = 7) received weekly intraperitoneal injections of viral particles carrying the sequence for Icon, resulting in expression of the protein, while the control group (n = 8) received no treatment. Icon preferentially reduced the number and volume of red vascularized lesions. Icon may present a novel treatment for endometriosis by degrading red vascularized lesions, likely by targeting tissue factor aberrantly expressed in the lesion vasculature.


Assuntos
Doenças dos Anexos/terapia , Endometriose/terapia , Fator VII/genética , Imunoconjugados/administração & dosagem , Fragmentos Fc das Imunoglobulinas/genética , Neovascularização Patológica/prevenção & controle , Proteínas Recombinantes de Fusão/genética , Tromboplastina/antagonistas & inibidores , Adenoviridae , Doenças dos Anexos/imunologia , Doenças dos Anexos/metabolismo , Doenças dos Anexos/patologia , Substituição de Aminoácidos , Animais , Endometriose/imunologia , Endometriose/metabolismo , Endometriose/patologia , Feminino , Terapia Genética , Vetores Genéticos , Humanos , Imunoconjugados/genética , Imunoglobulina G/genética , Terapia de Alvo Molecular , Mutação , Neovascularização Patológica/etiologia , Papio anubis , Pelve , Fragmentos de Peptídeos/genética , Distribuição Aleatória , Tromboplastina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA