Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Appl Immunohistochem Mol Morphol ; 30(4): 237-245, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35384873

RESUMO

The objectives were to develop a standardized Ki-67 immunohistochemistry (IHC) method for precise, robust, and reproducible assessment of patients with early breast cancer, and utilize this assay to evaluate patients participating in the monarchE study (NCT03155997). The Ki-67 assay was developed and validated for sensitivity, specificity, repeatability, precision, and robustness using a predefined ≥20% cutoff. Reproducibility studies (intersite and intrasite, interobserver and intraobserver) were conducted at 3 external laboratories using detailed scoring instructions designed for monarchE. Using the assay, patient tumors were classified as displaying high (≥20%) or low (<20%) Ki-67 expression; Kaplan-Meier methods evaluated 2-year invasive disease-free survival rates for these 2 groups among patients treated with endocrine therapy (ET) alone. All analytical validation and reproducibility studies achieved point estimates of >90% for negative, positive, and overall percent agreement. Intersite reproducibility produced point estimate values of 94.7%, 100.0%, and 97.3%. External interobserver reproducibility produced point estimate values of 98.9%, 97.8%, and 98.3%. Among 1954 patients receiving ET alone, 986 (50.5%) had high and 968 (49.5%) had low Ki-67 expression. Patients with high Ki-67 had a clinically meaningful increased risk of developing invasive disease within 2 years compared with those with low Ki-67 [2-y invasive disease-free survival rate: 86.1% (95% confidence interval: 83.1%-88.7%) vs. 92.0% (95% confidence interval: 89.7%-93.9%), respectively]. This standardized Ki-67 methodology resulted in high concordance across multiple laboratories, and its use in the monarchE study prospectively demonstrated the prognostic value of Ki-67 IHC in HR+, HER2- early breast cancer with high-risk clinicopathologic features.


Assuntos
Neoplasias da Mama , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/patologia , Feminino , Humanos , Imuno-Histoquímica , Antígeno Ki-67/metabolismo , Recidiva Local de Neoplasia , Receptor ErbB-2/metabolismo , Reprodutibilidade dos Testes
2.
Mol Ther Methods Clin Dev ; 14: 261-269, 2019 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-31453264

RESUMO

Gene therapy holds great potential for conditions such as cardiovascular disease, including atherosclerosis and also vascular cancers, yet available vectors such as the adeno-associated virus (rAAV) transduce the vasculature poorly. To enable retargeting, a single-chain antibody (scFv) that binds to the vascular cell-adhesion molecule (VCAM-1) overexpressed at areas of endothelial inflammation was site specifically and covalently conjugated to the exterior of rAAV6. To achieve conjugation, the scFv was functionalized with an orthogonal click chemistry group. This conjugation utilized site-specific sortase A methodology, thus preserving scFv binding capacity to VCAM-1. The AAV6 was separately functionalized with 4-azidophenyl glyoxal (APGO) via covalent adducts to arginine residues in the capsid's heparin co-receptor binding region. APGO functionalization removed native tropism, greatly reducing rAAV6-GFP transduction into all cells tested, and the effect was similar to the inhibition seen in the presence of heparin. Utilizing the incorporated functionalizations, the scFv was then covalently conjugated to the exterior of rAAV6 via strain-promoted azide-alkyne cycloaddition (SPAAC). With both the removal of native heparin tropism and the addition of VCAM-1 targeting, rAAV6 transduction of endothelial cells was greatly enhanced compared to control cells. Thus, this novel and modular targeting system could have further application in re-directing AAV6 toward inflamed endothelium for therapeutic use.

3.
J Control Release ; 226: 229-37, 2016 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-26855052

RESUMO

Endothelial cells (EC) represent an important target for pharmacologic intervention, given their central role in a wide variety of human pathophysiologic processes. Studies in lab animal species have established that conjugation of drugs and carriers with antibodies directed to surface targets like the Platelet Endothelial Cell Adhesion Molecule-1 (PECAM-1, a highly expressed endothelial transmembrane protein) help to achieve specific therapeutic interventions in ECs. To translate such "vascular immunotargeting" to clinical practice, it is necessary to replace antibodies by advanced ligands that are more amenable to use in humans. We report the molecular design of a single chain variable antibody fragment (scFv) that binds with high affinity to human PECAM-1 and cross-reacts with its counterpart in rats and other animal species, allowing parallel testing in vivo and in human endothelial cells in microfluidic model. Site-specific modification of the scFv allows conjugation of protein cargo and liposomes, enabling their endothelial targeting in these models. This study provides a template for molecular engineering of ligands, enabling studies of drug targeting in animal species and subsequent use in humans.


Assuntos
Sistemas de Liberação de Medicamentos , Endotélio/imunologia , Lipossomos/imunologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/imunologia , Anticorpos de Cadeia Única/imunologia , Sequência de Aminoácidos , Animais , Afinidade de Anticorpos , Linhagem Celular , Células Endoteliais/imunologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Lipossomos/administração & dosagem , Lipossomos/química , Lipossomos/farmacocinética , Ratos , Anticorpos de Cadeia Única/administração & dosagem , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/farmacocinética
4.
Nano Lett ; 15(1): 469-75, 2015 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-25442878

RESUMO

The size- and fluorescence-based sorting of micro- and nanoscale particles suspended in fluid presents a significant and important challenge for both sample analysis and for manufacturing of nanoparticle-based products. Here, we demonstrate a disposable microfluidic particle sorter that enables high-throughput, on-demand counting and binary sorting of submicron particles and cells using either fluorescence or an electrically based determination of particle size. Size-based sorting uses a resistive pulse sensor integrated on-chip, whereas fluorescence-based discrimination is achieved using on-the-fly optical image capture and analysis. Following detection and analysis, the individual particles are deflected using a pair of piezoelectric actuators, directing the particles into one of two desired output channels; the main flow goes into a third waste channel. The integrated system can achieve sorting fidelities of better than 98%, and the mechanism can successfully count and actuate, on demand, more than 60,000 particles/min.

5.
ACS Nano ; 7(6): 4933-45, 2013 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-23706031

RESUMO

Nanoparticles have opened new exciting avenues for both diagnostic and therapeutic applications in human disease, and targeted nanoparticles are increasingly used as specific drug delivery vehicles. The precise quantification of nanoparticle internalization is of importance to measure the impact of physical and chemical properties on the uptake of nanoparticles into target cells or into cells responsible for rapid clearance. Internalization of nanoparticles has been measured by various techniques, but comparability of data between different laboratories is impeded by lack of a generally accepted standardized assay. Furthermore, the distinction between associated and internalized particles has been a challenge for many years, although this distinction is critical for most research questions. Previously used methods to verify intracellular location are typically not quantitative and do not lend themselves to high-throughput analysis. Here, we developed a mathematical model which integrates the data from high-throughput flow cytometry measurements with data from quantitative confocal microscopy. The generic method described here will be a useful tool in biomedical nanotechnology studies. The method was then applied to measure the impact of surface coatings of vesosomes on their internalization by cells of the reticuloendothelial system (RES). RES cells are responsible for rapid clearance of nanoparticles, and the resulting fast blood clearance is one of the major challenges in biomedical applications of nanoparticles. Coating of vesosomes with long chain polyethylene glycol showed a trend for lower internalization by RES cells.


Assuntos
Nanopartículas , Poliestirenos/química , Poliestirenos/metabolismo , Animais , Transporte Biológico , Linhagem Celular , Humanos , Macrófagos/metabolismo , Camundongos , Sistema Fagocitário Mononuclear/citologia
6.
Mol Cancer Ther ; 8(5): 1312-8, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19417151

RESUMO

Cancer heterogeneity renders risk stratification and therapy decisions challenging. Thus, genomic and proteomic methodologies have been used in an effort to identify biomarkers that can differentiate tumor subtypes to improve therapeutic outcome. Here, we report a generally applicable strategy to generate tumor type-specific peptide ligand arrays. Peptides that specifically recognize breast tumor-derived cell lines (MDA-MB-231, MCF-7, and T47-D) were identified using cell-displayed peptide libraries carrying an intrinsic fluorescent marker allowing for sorting and characterization with quantitative flow cytometry. Tumor cell specificity was achieved by depleting libraries of ligands binding to normal mammary epithelial cells (HMEC and MCF-10A). Although integrin binding RGD motifs were favored by some cell lines, screening with RGD competitors yielded several novel consensus motifs exhibiting improved tumor specificity. The resultant peptide array contained multiple consensus motifs exhibiting strong similarity to breast tumor-associated proteins. Profiling a panel of breast cancer cell lines with the peptide array revealed receptor expression patterns distinctive for luminal or basal tumor subtypes. In addition, peptide displaying bacteria and peptide functionalized microparticles enabled fluorescent labeling of tumor cells and frozen tumor tissue sections. Our results indicate that cell surface profiling using highly specific breast tumor cell binding ligands may provide an efficient route for tumor subtype classification, biomarker identification, and for the development of targeted diagnostics and therapeutics.


Assuntos
Neoplasias da Mama/classificação , Neoplasias da Mama/genética , Perfilação da Expressão Gênica , Ligantes , Biblioteca de Peptídeos , Receptores de Superfície Celular/genética , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sítios de Ligação , Biomarcadores Tumorais , Linhagem Celular Transformada , Linhagem Celular Tumoral , Biologia Computacional , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos SCID
7.
Am J Respir Crit Care Med ; 180(3): 247-56, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19342415

RESUMO

RATIONALE: Endothelial thrombomodulin (TM) regulates thrombosis and inflammation. Diverse forms of pulmonary and vascular injury are accompanied by down-regulation of TM, which aggravates tissue injury. We postulated that anchoring TM to the endothelial surface would restore its protective functions. OBJECTIVES: To design an effective and safe strategy to treat pulmonary thrombotic and inflammatory injury. METHODS: We synthesized a fusion protein, designated scFv/TM, by linking the extracellular domain of mouse TM to a single-chain variable fragment of an antibody to platelet endothelial cell adhesion molecule-1 (PECAM-1). The targeting and protective functions of scFv/TM were tested in mouse models of lung ischemia-reperfusion and acute lung injury (ALI) caused by intratracheal endotoxin and hyperoxia, both of which caused approximately 50% reduction in the endogenous expression of TM. MEASUREMENTS AND MAIN RESULTS: Biochemical assays showed that scFv/TM accelerated protein C activation by thrombin and bound mouse PECAM-1 and cytokine high mobility group-B1. After intravenous injection, scFv/TM preferentially accumulated in the mouse pulmonary vasculature. In a lung model of ischemia-reperfusion injury, scFv/TM attenuated elevation of early growth response-1, inhibited pulmonary deposition of fibrin and leukocyte infiltration, and preserved blood oxygenation more effectively than soluble TM. In an ALI model, scFv/TM, but not soluble TM, suppressed activation of nuclear factor-kappaB, inflammation and edema in the lung and reduced mortality without causing hemorrhage. CONCLUSIONS: Targeting TM to the endothelium using an scFv anchor enhances its antithrombotic and antiinflammatory effectiveness in models of ALI.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Endotélio/metabolismo , Inflamação/metabolismo , Embolia Pulmonar/metabolismo , Mucosa Respiratória/metabolismo , Trombomodulina/metabolismo , Lesão Pulmonar Aguda/etiologia , Animais , Modelos Animais de Doenças , Endotélio/patologia , Inflamação/patologia , Proteínas de Fusão de Membrana , Camundongos , Camundongos Endogâmicos BALB C , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Proteína C/metabolismo , Embolia Pulmonar/complicações , Mucosa Respiratória/patologia , Trombomodulina/genética
8.
Biochim Biophys Acta ; 1778(4): 854-63, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18211818

RESUMO

Targeting the tumor vasculature and selectively modifying endothelial functions is an attractive anti-tumor strategy. We prepared polyethyleneglycol modified immunoliposomes (IL) directed against vascular cell adhesion molecule 1 (VCAM-1), a surface receptor over-expressed on tumor vessels, and investigated the liposomal targetability in vitro and in vivo. In vitro, anti-VCAM-1 liposomes displayed specific binding to activated endothelial cells under static conditions, as well as under simulated blood flow conditions. The in vivo targeting of IL was analysed in mice bearing human Colo 677 tumor xenografts 30 min and 24 h post i.v. injection. Whereas biodistribution studies using [3H]-labelled liposomes displayed only marginal higher tumor accumulation of VCAM-1 targeted versus unspecific ILs, fluorescence microscopy evaluation revealed that their localisations within tumors differed strongly. VCAM-1 targeted ILs accumulated in tumor vessels with increasing intensities from 30 min to 24 h, while control ILs accumulated in the tumor tissue by passive diffusion. ILs that accumulated in non-affected organs, mainly liver and spleen, primarily co-localised with macrophages. This is the first morphological evidence for selective in vivo targeting of tumor vessels using ILs. VCAM-directed ILs are candidate drug delivery systems for therapeutic anti-cancer approaches designed to alter endothelial function.


Assuntos
Neoplasias/irrigação sanguínea , Molécula 1 de Adesão de Célula Vascular/metabolismo , Animais , Linhagem Celular Tumoral , Células Endoteliais/patologia , Feminino , Humanos , Lipossomos , Camundongos , Camundongos Nus , Especificidade de Órgãos , Tamanho da Partícula , Rodaminas
9.
Blood ; 111(4): 1999-2006, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18045968

RESUMO

A recombinant prodrug, single-chain urokinase-type plasminogen activator (scuPA) fused to an anti-PECAM-1 antibody single-chain variable fragment (anti-PECAM scFv/scuPA) targets endothelium and augments thrombolysis in the pulmonary vasculature.(1) To avoid premature activation and inactivation and to limit systemic toxicity, we replaced the native plasmin activation site in scFv/low-molecular-weight (lmw)-scuPA with a thrombin activation site, generating anti-PECAM scFv/uPA-T that (1) is latent and activated by thrombin instead of plasmin; (2) binds to PECAM-1; (3) does not consume plasma fibrinogen; (4) accumulates in mouse lungs after intravenous injection; and (5) resists PA inhibitor PAI-1 until activated by thrombin. In mouse models of pulmonary thrombosis caused by thromboplastin and ischemia-reperfusion (I/R), scFv/uPA-T provided more potent thromboprophylaxis and greater lung protection than plasmin-sensitive scFv/uPA. Endothelium-targeted thromboprophylaxis triggered by a prothrombotic enzyme illustrates a novel approach to time- and site-specific regulation of proteolytic reactions that can be modulated for therapeutic benefit.


Assuntos
Endotélio Vascular/fisiologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/fisiologia , Circulação Pulmonar/fisiologia , Trombina/fisiologia , Terapia Trombolítica , Ativador de Plasminogênio Tipo Uroquinase/fisiologia , Animais , Endotélio Vascular/efeitos dos fármacos , Fibrinogênio/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Ligação Proteica , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Distribuição Tecidual , Ativador de Plasminogênio Tipo Uroquinase/genética , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
10.
J Pharmacol Exp Ther ; 321(3): 947-52, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17389242

RESUMO

Efficacy and safety of current means to prevent cerebrovascular thrombosis in patients at high risk of stroke are suboptimal. In theory, anchoring fibrinolytic plasminogen activators to the luminal surface of the cerebral endothelium might arrest formation of occlusive clots in this setting. We tested this approach using the recombinant construct antiplatelet-endothelial cell adhesion molecule (PECAM) single-chain variable fragment (scFv)-urokinase-type plasminogen activator (uPA), fusing low-molecular-weight single-chain urokinase-type plasminogen activator with a scFv of an antibody directed to the stably expressed endothelial surface determinant PECAM-1, implicated in inflammation and thrombosis. Studies in mice showed that scFv-uPA, but not unconjugated uPA 1) accumulates in the brain after intravascular injection, 2) lyses clots lodged in the cerebral arterial vasculature without hemorrhagic complications, 3) provides rapid and stable cerebral reperfusion, and 4) alleviates post-thrombotic brain edema. Effective and safe thromboprophylaxis in the cerebral arterial circulation by anti-PECAM scFv-uPA represents a prototype of a new paradigm to prevent recurrent cerebrovascular thrombosis.


Assuntos
Edema Encefálico/prevenção & controle , Sistemas de Liberação de Medicamentos/métodos , Fragmentos de Imunoglobulinas/imunologia , Trombose Intracraniana/prevenção & controle , Molécula-1 de Adesão Celular Endotelial a Plaquetas/imunologia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Encéfalo/irrigação sanguínea , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Química Encefálica/efeitos dos fármacos , Edema Encefálico/patologia , Cerebelo/efeitos dos fármacos , Cerebelo/metabolismo , Cerebelo/patologia , Circulação Cerebrovascular/efeitos dos fármacos , Fibrinólise/efeitos dos fármacos , Fragmentos de Imunoglobulinas/genética , Injeções Intra-Arteriais , Trombose Intracraniana/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pró-Fármacos/administração & dosagem , Pró-Fármacos/uso terapêutico , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética , Lobo Temporal/efeitos dos fármacos , Lobo Temporal/metabolismo , Lobo Temporal/patologia , Terapia Trombolítica/métodos , Ativador de Plasminogênio Tipo Uroquinase/genética
11.
Thromb J ; 3: 21, 2005 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-16336690

RESUMO

BACKGROUND: Vascular targeting of malignant tumors has become a clinically validated new treatment approach with clear patient benefit. However clinical studies have also revealed that some types of vascular targeting agents (VTAs) are prone to coagulation system side effects. It is therefore essential to predetermine coagulation parameters in preclinical studies. As of to date, this has rarely been done, predominantly due to technical issues. The goal of this study was to establish and apply a standardized process, whereby systemic coagulation activation can be routinely measured in mice. RESULTS: We have evaluated a number of sampling techniques and coagulation tests regarding their suitability for this purpose. We were able to adapt two assays measuring soluble fibrin, a marker for a prethrombotic status. Thus, soluble fibrin could be measured for the first time in mice. All assays were validated in a positive control model for systemic coagulation activation, i.e. lipopolysaccharide-induced endotoxemia. Based on our results, we selected a panel of coagulation tests, which are both feasable and informative for preclinical testing of VTAs: soluble fibrin, thrombin-antithrombin complexes, free antithrombin III, white blood cell counts and platelet counts. The effect of tumor transplants on coagulation parameters was evaluated using this panel. We then applied this set of assays in treatment studies with a VTA developed in our laboratory to investigate a potential systemic coagulation activation. CONCLUSION: We have established a standardized panel of assays that can be used to test murine blood samples for coagulation activation in preclinical studies. All tests are feasible to perform in any research laboratory without specialized equipment. In addition, this is the first report to measure soluble fibrin, an early marker of systemic coagulation activation, in mice. The panel was applied on tumor bearing mice and mice treated with a VTA. We suggest its general application for coagulation activation analyses in mice.

12.
Blood ; 106(13): 4191-8, 2005 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-16144802

RESUMO

Means to prevent thrombus extension and local recurrence remain suboptimal, in part because of the limited effectiveness of existing thrombolytics. In theory, plasminogen activators could be used for this purpose if they could be anchored to the vascular lumen by targeting stably expressed, noninternalized determinants such as platelet-endothelial-cell adhesion molecule 1 (PECAM-1). We designed a recombinant molecule fusing low-molecular-weight single-chain prourokinase plasminogen activator (lmw-scuPA) with a single-chain variable fragment (scFv) of a PECAM-1 antibody to generate the prodrug scFv/lmw-scuPA. Cleavage by plasmin generated fibrinolytically active 2-chain lmw-uPA. This fusion protein (1) bound specifically to PECAM-1-expressing cells; (2) was rapidly cleared from blood after intravenous injection; (3) accumulated in the lungs of wild-type C57BL6/J, but not PECAM-1 null mice; and (4) lysed pulmonary emboli formed subsequently more effectively than lmw-scuPA, thereby providing support for the concept of thromboprophylaxis using recombinant scFv-fibrinolytic fusion proteins that target endothelium.


Assuntos
Células Endoteliais/metabolismo , Fragmentos de Imunoglobulinas/metabolismo , Pulmão/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Trombose/metabolismo , Trombose/patologia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Linhagem Celular , Drosophila , Células Endoteliais/patologia , Humanos , Fragmentos de Imunoglobulinas/genética , Pulmão/irrigação sanguínea , Pulmão/patologia , Camundongos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/genética
13.
J Natl Cancer Inst ; 97(10): 733-47, 2005 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-15900043

RESUMO

BACKGROUND: The tumor vasculature is increasingly recognized as a target for cancer therapy. We developed and evaluated recombinant fusion proteins targeting the coagulation-inducing protein soluble tissue factor (sTF) to the luminal tumor endothelial antigen vascular cell adhesion molecule 1 (VCAM-1, CD106). METHODS: We generated fusion proteins consisting of sTF fused to antibody fragments directed against mouse or human VCAM-1 and characterized them in vitro by flow cytometry, surface plasmon resonance, and two-stage coagulation assays. Their therapeutic effects were tested in three human xenograft tumor models: L540rec Hodgkin lymphoma, Colo677 small-cell lung carcinoma, and Colo677/HDMEC small-cell lung carcinoma with human vasculature. Toxicity was analyzed by histologic examination of organs and determination of laboratory blood parameters. RESULTS: The fusion proteins bound VCAM-1 with nanomolar affinities and had the same coagulation activity as an sTF standard. Xenograft tumor-bearing mice treated with fusion protein (FP) alone or in combination with lipopolysaccharide (FP/L) or doxorubicin (FP/D) exhibited tumor-selective necrosis (L540rec tumors: 74% tumor necrosis [95% confidence interval {CI} = 55% to 93%] with FP/L versus 13% tumor necrosis [95% CI = 4% to 22%] with vehicle; Colo677 tumors: 26% [95% CI = 16% to 36%] with FP versus 8% [95% CI = 2% to 14%] with vehicle); tumor growth delay (Colo677/HDMEC: mean tumor weights after 3 days = 42 mg in FP-treated mice versus 71 mg in vehicle-treated mice, difference = 29 mg, 95% CI = 8 to 100, Mann-Whitney P = .008); and some tumor regressions (one of seven FP-treated Colo677 tumor-bearing mice and two of seven FP/D-treated mice). The fusion protein was well tolerated. CONCLUSIONS: Recombinant tissue factor-based fusion proteins directed against an intraluminal tumor endothelial cell marker induce tumor-selective intravascular coagulation, tumor tissue necrosis, and tumor growth delay.


Assuntos
Inibidores da Angiogênese/farmacologia , Neoplasias Experimentais/irrigação sanguínea , Neoplasias Experimentais/tratamento farmacológico , Proteínas Recombinantes de Fusão/farmacologia , Molécula 1 de Adesão de Célula Vascular/efeitos dos fármacos , Inibidores da Angiogênese/química , Inibidores da Angiogênese/uso terapêutico , Animais , Testes de Coagulação Sanguínea , Carcinoma de Células Pequenas/irrigação sanguínea , Carcinoma de Células Pequenas/tratamento farmacológico , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Doença de Hodgkin/tratamento farmacológico , Doença de Hodgkin/patologia , Humanos , Imuno-Histoquímica , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/tratamento farmacológico , Camundongos , Microcirculação/efeitos dos fármacos , Necrose , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/uso terapêutico , Projetos de Pesquisa , Ressonância de Plasmônio de Superfície , Transplante Heterólogo
14.
Blood ; 102(10): 3737-42, 2003 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-12881320

RESUMO

CD30 is a promising target for antibody-based immunotherapy of Hodgkin lymphoma (HL) and anaplastic large cell lymphoma. To overcome the limitations from currently available murine anti-CD30 monoclonal antibodies (mAbs), a new fully human anti-CD30 antibody was generated. Binding properties were evaluated by recombinant CD30 capture enzyme-linked immunosorbent assay (ELISA) and fluorescence-activated cell-sorter (FACS) flow cytometry. Activity of this new mAb was assessed in vitro using growth inhibition and antibody-dependent cellular cytotoxicity (ADCC) assays on several cell lines. In vivo activity was determined in a solid as well as in a disseminated xenografted model of HL in severe combined immunodeficiency (SCID) mice. The mAb 5F11 showed specific binding to CD30 (cluster A). The ADCC assays indicated dose-dependent lysis of L540 cells when 5F11 was combined with human effector cells. Upon cross-linking in vitro, 5F11 inhibited the growth of CD30-expressing cell lines. In vivo, treatment with 5F11 induced a marked growth delay or even a complete regression of established xenografted HL in SCID mice. In the disseminated HL model, a high proportion of 5F11-treated mice experienced long-term survival. The new human anti-CD30 monoclonal antibody 5F11 shows promise as a means of CD30-targeted immunotherapy of malignant lymphomas. Based on these results, a clinical phase 1 study in patients with refractory CD30+ lymphoma has been initiated.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antígeno Ki-1/imunologia , Linfoma/tratamento farmacológico , Animais , Anticorpos Monoclonais/farmacologia , Afinidade de Anticorpos , Antígenos de Neoplasias/imunologia , Divisão Celular , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Feminino , Doença de Hodgkin/tratamento farmacológico , Doença de Hodgkin/mortalidade , Humanos , Linfoma/mortalidade , Camundongos , Camundongos SCID , Transplante de Neoplasias , Taxa de Sobrevida , Transplante Heterólogo
15.
Arterioscler Thromb Vasc Biol ; 23(5): 905-10, 2003 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-12649087

RESUMO

OBJECTIVE: This study was performed to evaluate the mechanisms leading to tumor vessel occlusion by tissue factor-based drugs, which are used in vascular targeting approaches for the treatment of malignant tumors. METHODS AND RESULTS: The effects of nontargeted soluble tissue factor were evaluated in vitro and in vivo. Tumor-bearing mice were treated with (1) the extracellular portion of tissue factor (soluble tissue factor), (2) low nontoxic doses of lipopolysaccharides, or (3) a combination thereof. The combination treatment showed the best effects and resulted in selective thrombosis of tumor vessels. On the basis of our data from subsequent in vitro analyses, including surface plasmon resonance measurements and endothelial cell based coagulation assays, we propose a model on how soluble tissue factor, although lacking its membrane anchor, can promote selective tumor vessel occlusion. CONCLUSIONS: To our knowledge, this is the first report to describe the molecular mechanisms of coagulation induction by untargeted soluble tissue factor in vivo. Combination treatments including soluble tissue factor might represent an alternative vascular targeting approach for the treatment of malignant tumors.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Embolização Terapêutica/métodos , Lipopolissacarídeos/uso terapêutico , Linfoma não Hodgkin/terapia , Teratocarcinoma/terapia , Tromboplastina/uso terapêutico , Animais , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Quimioterapia Combinada , Ativação Enzimática/efeitos dos fármacos , Fator Xa/biossíntese , Humanos , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/farmacologia , Linfoma não Hodgkin/sangue , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/farmacologia , Fragmentos de Peptídeos/uso terapêutico , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Solubilidade , Ressonância de Plasmônio de Superfície , Teratocarcinoma/irrigação sanguínea , Tromboplastina/administração & dosagem , Tromboplastina/genética , Tromboplastina/farmacologia , Fator de Necrose Tumoral alfa/análise , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...