Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Trends Biochem Sci ; 43(1): 18-31, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29162390

RESUMO

Heparanase, the sole heparan sulfate (HS)-degrading endoglycosidase, regulates multiple biological activities that enhance tumor growth, metastasis, angiogenesis, and inflammation. Heparanase accomplishes this by degrading HS and thereby regulating the bioavailability of heparin-binding proteins; priming the tumor microenvironment; mediating tumor-host crosstalk; and inducing gene transcription, signaling pathways, exosome formation, and autophagy that together promote tumor cell performance and chemoresistance. By contrast, heparanase-2, a close homolog of heparanase, lacks enzymatic activity, inhibits heparanase activity, and regulates selected genes that promote normal differentiation, endoplasmic reticulum stress, tumor fibrosis, and apoptosis, together resulting in tumor suppression. The emerging premise is that heparanase is a master regulator of the aggressive phenotype of cancer, while heparanase-2 functions as a tumor suppressor.


Assuntos
Neoplasias/metabolismo , Neoplasias/patologia , Polissacarídeo-Liases/metabolismo , Animais , Progressão da Doença , Humanos
2.
Proc Natl Acad Sci U S A ; 113(3): 704-9, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26729870

RESUMO

Heparanase is an endoglycosidase that cleaves heparan sulfate side chains of proteoglycans, resulting in disassembly of the extracellular matrix underlying endothelial and epithelial cells and associating with enhanced cell invasion and metastasis. Heparanase expression is induced in carcinomas and sarcomas, often associating with enhanced tumor metastasis and poor prognosis. In contrast, the function of heparanase in hematological malignancies (except myeloma) was not investigated in depth. Here, we provide evidence that heparanase is expressed by human follicular and diffused non-Hodgkin's B-lymphomas, and that heparanase inhibitors restrain the growth of tumor xenografts produced by lymphoma cell lines. Furthermore, we describe, for the first time to our knowledge, the development and characterization of heparanase-neutralizing monoclonal antibodies that inhibit cell invasion and tumor metastasis, the hallmark of heparanase activity. Using luciferase-labeled Raji lymphoma cells, we show that the heparanase-neutralizing monoclonal antibodies profoundly inhibit tumor load in the mouse bones, associating with reduced cell proliferation and angiogenesis. Notably, we found that Raji cells lack intrinsic heparanase activity, but tumor xenografts produced by this cell line exhibit typical heparanase activity, likely contributed by host cells composing the tumor microenvironment. Thus, the neutralizing monoclonal antibodies attenuate lymphoma growth by targeting heparanase in the tumor microenvironment.


Assuntos
Anticorpos Neutralizantes/farmacologia , Anticorpos Neutralizantes/uso terapêutico , Glucuronidase/imunologia , Linfoma/patologia , Animais , Anticorpos Monoclonais/farmacologia , Proliferação de Células/efeitos dos fármacos , Glucuronidase/isolamento & purificação , Células HEK293 , Humanos , Luciferases/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos SCID , Peso Molecular , Metástase Neoplásica , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Saponinas/farmacologia , Carga Tumoral/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Matrix Biol ; 77: 58-72, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30096360

RESUMO

It is now well recognized that heparanase, an endo-ß-D-glucuronidase capable of cleaving heparan sulfate (HS) side chains at a limited number of sites, promotes tumorigenesis by diverse mechanisms. Compelling evidence strongly implies that heparanase is a viable target for cancer therapy, thus encouraging the development of heparanase inhibitors as anti-cancer therapeutics. Here, we examined the efficacy and mode of action of PG545, an HS-mimetic heparanase inhibitor, in human lymphoma. We found that PG545 exhibits a strong anti-lymphoma effect, eliciting lymphoma cell apoptosis. Notably, this anti-lymphoma effect involves ER stress response that was accompanied by increased autophagy. The persistent ER stress evoked by PG545 is held responsible for cell apoptosis because apoptotic cell death was attenuated by an inhibitor of PERK, a molecular effector of ER stress. Importantly, PG545 had no such apoptotic effect on naïve splenocytes, further encouraging the development of this compound as anti-lymphoma drug. Surprisingly, we found that PG545 also elicits apoptosis in lymphoma cells that are devoid of heparanase activity (i.e., Raji), indicating that the drug also exerts heparanase-independent function(s) that together underlie the high potency of PG545 in preclinical cancer models.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Carcinogênese/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Glucuronidase/genética , Linfoma/tratamento farmacológico , Saponinas/farmacologia , Animais , Anticorpos Monoclonais/farmacologia , Apoptose/genética , Autofagia/efeitos dos fármacos , Autofagia/genética , Carcinogênese/genética , Carcinogênese/metabolismo , Carcinogênese/patologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/genética , Linhagem Celular Tumoral , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Estresse do Retículo Endoplasmático/genética , Regulação da Expressão Gênica , Glucuronidase/antagonistas & inibidores , Glucuronidase/metabolismo , Heparina/análogos & derivados , Heparina/farmacologia , Heparitina Sulfato , Humanos , Linfócitos/citologia , Linfócitos/efeitos dos fármacos , Linfócitos/enzimologia , Linfoma/enzimologia , Linfoma/genética , Linfoma/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Cultura Primária de Células , Baço/citologia , Baço/efeitos dos fármacos , Baço/enzimologia , Ensaios Antitumorais Modelo de Xenoenxerto
4.
ACS Chem Biol ; 13(12): 3236-3242, 2018 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-30480427

RESUMO

Heparanase is a mammalian endoglycosidase that cleaves heparan sulfate (HS) polysaccharides and contributes to remodelling of the extracellular matrix and regulation of HS-binding protein bioavailabilities. Heparanase is upregulated in malignant cancers and inflammation, aiding cell migration and the release of signaling molecules. It is established as a highly druggable extracellular target for anticancer therapy, but current compounds have limitations, because of cost, production complexity, or off-target effects. Here, we report the synthesis of a novel, targeted library of single-entity glycomimetic clusters capped with simple sulfated saccharides. Several dendrimer HS glycomimetics display low nM IC50 potency for heparanase inhibition equivalent to comparator compounds in clinical development, and potently inhibit metastasis and growth of human myeloma tumor cells in a mouse xenograft model. Importantly, they lack anticoagulant activity and cytotoxicity, and also inhibit angiogenesis. They provide a new candidate class for anticancer and wider therapeutic applications, which could benefit from targeted heparanase inhibition.


Assuntos
Antineoplásicos/uso terapêutico , Materiais Biomiméticos/uso terapêutico , Dendrímeros/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Glucuronidase/antagonistas & inibidores , Mieloma Múltiplo/tratamento farmacológico , Inibidores da Angiogênese/síntese química , Inibidores da Angiogênese/farmacologia , Inibidores da Angiogênese/uso terapêutico , Inibidores da Angiogênese/toxicidade , Animais , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Antineoplásicos/toxicidade , Materiais Biomiméticos/síntese química , Materiais Biomiméticos/farmacologia , Materiais Biomiméticos/toxicidade , Linhagem Celular Tumoral , Dendrímeros/síntese química , Dendrímeros/farmacologia , Dendrímeros/toxicidade , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/toxicidade , Fator 2 de Crescimento de Fibroblastos/antagonistas & inibidores , Glicosídeos/síntese química , Glicosídeos/farmacologia , Glicosídeos/uso terapêutico , Glicosídeos/toxicidade , Heparitina Sulfato/química , Humanos , Concentração Inibidora 50 , Camundongos , Estrutura Molecular , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Hum Vaccin Immunother ; 12(9): 2253-6, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27054564

RESUMO

Heparanase, the sole heparan sulfate degrading endoglycosidase, regulates multiple biological activities that enhance tumor growth, angiogenesis and metastasis. Much of the impact of heparanase on tumor progression is related to its function in mediating tumor-host crosstalk, priming the tumor microenvironment to better support tumor progression. Heparanase expression is enhanced in almost all cancers examined including various carcinomas, sarcomas and hematological malignancies. Numerous clinical association studies have consistently demonstrated that upregulated heparanase expression correlates with increased tumor size, tumor angiogenesis, enhanced metastasis and poor prognosis. Notably, heparanase is ranked among the most frequently recognized tumor antigens in patients with pancreatic, colorectal or breast cancer, favoring heparanase-based immunotherapy. Development of heparanase inhibitors focused on carbohydrate-based compounds of which 4 are being evaluated in clinical trials for various types of cancer, including myeloma, pancreatic carcinoma and hepatocellular carcinoma. Owing to their heparin-like nature, these compounds may exert off target effects. Newly generated heparanase neutralizing monoclonal antibodies profoundly attenuated myeloma and lymphoma tumor growth and dissemination in preclinical models, likely by targeting heparanase in the tumor microenvironment.


Assuntos
Carcinogênese , Glucuronidase/metabolismo , Neoplasias/fisiopatologia , Animais , Antineoplásicos/uso terapêutico , Ensaios Clínicos como Assunto , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/uso terapêutico , Glucuronidase/antagonistas & inibidores , Humanos
6.
Oncotarget ; 7(44): 71362-71377, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27655685

RESUMO

Recurrence of breast cancer disease years after treatment appears to arise from disseminated dormant tumor cells (DTC). The mechanisms underlying the outgrowth of DTC remain largely unknown. Here we demonstrate that dormant MCF-7 cells expressing LOXL2 acquire a cancer stem cell (CSC)-like phenotype, mediating their outgrowth in the 3D BME system that models tumor dormancy and outgrowth. Similarly, MCF-7-LOXL2 cells colonizing the lung transitioned from dormancy to metastatic outgrowth whereas MCF-7 cells remained dormant. Notably, epithelial to mesenchymal transition (EMT) of MCF-7-LOXL2 cells was required for their CSC-like properties and their transition to metastatic outgrowth. These findings were further supported by clinical data demonstrating that increase in LOXL2 mRNA levels correlates with increase in the mRNA levels of EMT and stem cells markers, and is also associated with decrease in relapse free survival of breast cancer patients. Notably, conditional hypoxia induced expression of endogenous LOXL2 in MCF-7 cells promoted EMT and the acquisition of a CSC-like phenotype, while knockdown of LOXL2 inhibited this transition. Overall, our results demonstrate that expression of LOXL2 endowed DTC with CSC-like phenotype driving their transition to metastatic outgrowth and this stem-like phenotype is dependent on EMT that can be driven by the tumor microenvironment.


Assuntos
Aminoácido Oxirredutases/fisiologia , Neoplasias da Mama/patologia , Células-Tronco Neoplásicas/fisiologia , Aminoácido Oxirredutases/genética , Neoplasias da Mama/mortalidade , Hipóxia Celular , Proliferação de Células , Transição Epitelial-Mesenquimal , Feminino , Humanos , Células MCF-7 , Metástase Neoplásica , Recidiva Local de Neoplasia , Fenótipo , Microambiente Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA