Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Biomed Pharmacother ; 110: 803-817, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30554119

RESUMO

Cancer stem cells (CSCs) are the promising targets for cancer chemotherapy that cannot be eliminated by conventional chemotherapy. In this study cationic liposomes of cabazitaxel (CBX) and silibinin (SIL) were prepared with an aim to kill cancer cells and CSCs for prostate cancer. CBX act as cancer cell inhibitor and SIL as CSC inhibitor. Hyaluronic acid (HA), an endogenous anionic polysaccharide was coated on cationic liposomes for targeting CD44 receptors over expressed on CSCs. Liposomes were prepared by ethanol injection method with particle size below 100 nm and entrapment efficiency of more than 90% at 10% w/w drug loading. Liposomes were characterized by dynamic light scattering, transmission electron microscopy, 1H nuclear magnetic resonance and scanning electron microscopy-energy dispersive x-ray spectroscopy. Liposomes were evaluated for their anticancer action in androgen independent human prostate cancer cell lines (PC-3 and DU-145). HA coated liposomes showed potential cytotoxicity over other groups with low IC50, significantly inhibited cell migration and induced apoptosis. Synergistic cytotoxic effect was also observed with HA coated liposomes that resulted in colony formation inhibition and G2/M phase arrest. Proficient cytotoxicity against CD44+ cells (14.87 ± 0.41% in PC-3 and 33.95 ± 0.68% in DU-145 cells) indicated the efficiency of HA coated liposomes towards CSC targeting. Hence, the outcome of this combinational therapy with CD44 targeting indicates the suitability of HA coated CBX and SIL co-loaded liposomes as a potential approach for eradicating prostate cancer and herein might provide a insight for future studies.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Receptores de Hialuronatos/administração & dosagem , Nanomedicina/métodos , Neoplasias da Próstata , Silibina/administração & dosagem , Taxoides/administração & dosagem , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Cátions , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Humanos , Receptores de Hialuronatos/metabolismo , Lipossomos , Masculino , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/metabolismo , Silibina/farmacocinética , Taxoides/farmacocinética , Ensaio Tumoral de Célula-Tronco/métodos
2.
Tumour Biol ; 39(5): 1010428317701628, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28468593

RESUMO

Peritoneal carcinomatosis results from dissemination of solid tumors in the peritoneal cavity, and is a common site of metastasis in patients with carcinomas of gastrointestinal or gynecological origin. Peritoneal carcinomatosis treatment is challenging as poorly vascularized, disseminated peritoneal micro-tumors are shielded from systemic anticancer drugs and drive tumor regrowth. Here, we describe the identification and validation of a tumor homing peptide CKRDLSRRC (IP3), which upon intraperitoneal administration delivers payloads to peritoneal metastases. IP3 peptide was identified by in vivo phage display on a mouse model of peritoneal carcinomatosis of gastric origin (MKN-45P), using high-throughput sequencing of the peptide-encoding region of phage genome as a readout. The IP3 peptide contains a hyaluronan-binding motif, and fluorescein-labeled IP3 peptide bound to immobilized hyaluronan in vitro. After intraperitoneal administration in mice bearing peritoneal metastases of gastric and colon origin, IP3 peptide homed robustly to macrophage-rich regions in peritoneal tumors, including poorly vascularized micro-tumors. Finally, we show that IP3 functionalization conferred silver nanoparticles the ability to home to peritoneal tumors of gastric and colonic origin, suggesting that it could facilitate targeted delivery of nanoscale payloads to peritoneal tumors. Collectively, our study suggests that the IP3 peptide has potential applications for targeting drugs, nanoparticles, and imaging agents to peritoneal tumors.


Assuntos
Carcinoma/tratamento farmacológico , Receptores de Hialuronatos/administração & dosagem , Peptídeos/administração & dosagem , Neoplasias Peritoneais/tratamento farmacológico , Animais , Bacteriófagos/genética , Carcinoma/genética , Carcinoma/patologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Humanos , Receptores de Hialuronatos/genética , Camundongos , Nanopartículas/administração & dosagem , Nanopartículas/química , Metástase Neoplásica , Peptídeos/genética , Cavidade Peritoneal/patologia , Neoplasias Peritoneais/genética , Neoplasias Peritoneais/patologia
3.
Cancer Sci ; 108(7): 1493-1503, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28444967

RESUMO

Tumor metastasis occurs naturally in pancreatic cancer, and the efficacy of chemotherapy is usually poor. Precision medicine, combining downregulation of target genes with chemotherapy drugs, is expected to improve therapeutic effects. Therefore, we developed a combined therapy of microRNA-21 antisense oligonucleotides (ASO-miR-21) and gemcitabine (Gem) using a targeted co-delivery nanoparticle (NP) carrier and investigated the synergistic inhibitory effects on pancreatic cancer cells metastasis and growth. Polyethylene glycol-polyethylenimine-magnetic iron oxide NPs were used to co-deliver ASO-miR-21 and Gem. An anti-CD44v6 single-chain variable fragment (scFvCD44v6 ) was used to coat the particles to obtain active and targeted delivery. Our results showed that the downregulation of the oncogenic miR-21 by ASO resulted in upregulation of the tumor-suppressor genes PDCD4 and PTEN and the suppression of epithelial-mesenchymal transition, which inhibited the proliferation and induced the clonal formation, migration, and invasion of pancreatic cancer cells in vitro. The co-delivery of ASO-miR-21 and Gem induced more cell apoptosis and inhibited the growth of pancreatic cancer cells to a greater extent than single ASO-miR-21 or Gem treatment in vitro. In animal tests, more scFvCD44v6 -PEG-polyethylenimine/ASO-magnetic iron oxide NP/Gem accumulated at the tumor site than non-targeted NPs and induced a potent inhibition of tumor proliferation and metastasis. Magnetic resonance imaging was used to observed tumor homing of NPs. These results imply that the combination of miR-21 gene silencing and Gem therapy using an scFv-functionalized NP carrier exerted synergistic antitumor effects on pancreatic cancer cells, which is a promising strategy for pancreatic cancer therapy.


Assuntos
Desoxicitidina/análogos & derivados , Terapia Genética/métodos , MicroRNAs/antagonistas & inibidores , Terapia de Alvo Molecular/métodos , Oligonucleotídeos Antissenso/administração & dosagem , Neoplasias Pancreáticas/patologia , Animais , Desoxicitidina/administração & dosagem , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/química , Feminino , Compostos Férricos , Humanos , Receptores de Hialuronatos/administração & dosagem , Nanopartículas Metálicas/química , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanomedicina/métodos , Polietilenoglicóis , Polietilenoimina , Medicina de Precisão/métodos , Anticorpos de Cadeia Única/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina
4.
Vopr Onkol ; 62(3): 514-8, 2016.
Artigo em Russo | MEDLINE | ID: mdl-30463110

RESUMO

In this work the effect of RHAMM (receptor hyaluronan-mediated motility)-target peptides was investigated on the viability, apoptosis and necrosis of prostate cancer cells (PC3m-LN4). It has been established that RHAMM-target peptides inhibited on 90 % cell viability of PC3m-LN4 cells at a concentration of 10 ug / ml (2х10-7 M) for 48 h. It has shown that RHAMM-target peptides induced apoptosis and inhibited necrosis of tumor cells. RHAMM-target peptide had no effect on fibroblasts (non-tumor cells) and fibroblasts (RHAMM-/-). The studies also revealed that RHAMM-target peptides enhanced activity of caspase-3/7 in cancer cells.


Assuntos
Proliferação de Células/efeitos dos fármacos , Proteínas da Matriz Extracelular/genética , Receptores de Hialuronatos/genética , Peptídeos/administração & dosagem , Neoplasias da Próstata/genética , Apoptose/efeitos dos fármacos , Caspase 3/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Proteínas da Matriz Extracelular/administração & dosagem , Fibroblastos/efeitos dos fármacos , Humanos , Receptores de Hialuronatos/administração & dosagem , Masculino , Peptídeos/genética , Neoplasias da Próstata/patologia , Neoplasias da Próstata/terapia , Transfecção
5.
BioDrugs ; 28(4): 393-402, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24567264

RESUMO

BACKGROUND AND OBJECTIVES: The aim of this study was to increase the serum half-life of recombinant CD44 hyaluronan (HA) binding domain by PEGylation. We have previously found that recombinant soluble CD44 HA binding domain (CD44HABD) and its non-HA-binding triple mutant CD44HABD(R41AY78SY79S) (CD44-3MUT) inhibits angiogenesis and subcutaneous tumor growth. However, this ~12 kDa recombinant protein displays a high serum clearance rate. METHODS: Here, we report the purification of monomeric CD44-3MUT from urea solubilized inclusion bodies using weak anion exchange chromatography and gel filtration. To increase the serum residence time of CD44-3MUT we PEGylated the resulting protein using 20 kDa methoxy-PEG-propionaldehyde. RESULTS: PEGylation of CD44-3MUT prolonged its in vivo serum half-life about 70-fold from 0.03 to 1.8 hours. Along with extended plasma residence time, PEGylation also increased the systemic exposure. By cell impedance assay we confirmed that PEGylated CD44-3MUT maintained its in vitro function. The results from the impedance assay additionally demonstrate that the CD44-3MUT effect on endothelial cells is mediated by vimentin. CONCLUSIONS: In summary, we have developed a purification protocol for large-scale production of CD44-3MUT and generated a PEGylated form of CD44-3MUT. HA binding domain of CD44(CD44HABD) and its modified non-HA binding form (CD44-3MUT) inhibit angiogenesis and tumor growth in vivo without disturbing HA-binding functions. CD44-3MUT has been PEGylated for use as a new type of anti-angiogenic human drug. PEGylation of CD44-3MUT improved pharmacokinetic properties but retains its functional activity.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Receptores de Hialuronatos/metabolismo , Ácido Hialurônico/metabolismo , Polietilenoglicóis/química , Inibidores da Angiogênese/farmacocinética , Inibidores da Angiogênese/farmacologia , Animais , Feminino , Meia-Vida , Receptores de Hialuronatos/administração & dosagem , Camundongos , Ligação Proteica , Ratos , Ratos Endogâmicos F344 , Proteínas Recombinantes/metabolismo , Vimentina/metabolismo
6.
J Drug Target ; 20(7): 561-73, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22758394

RESUMO

CD44 or hyaluronan receptor is a transmembrane receptor associated with aggressive tumour growth, proliferation, and metastasis. In normal physiology, this receptor has a crucial role in cell adhesion, inflammation, and repair processes. However, many tumour cells over-express this receptor and abuse it to become progressive and perpetual units. The article comments from common functioning of the CD44 receptor, to its diabolic multi-dimensional effects in promotion of malignant cells. It also illuminates the relations of CD44 endorsed processes with other biomolecular events in cancer progression. In an end, the review focuses comprehensively at ongoing researches to exploit the CD44 over-expression as a probable target in treatment, management, and diagnosis of malignancy.


Assuntos
Antineoplásicos/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Receptores de Hialuronatos/fisiologia , Neoplasias/fisiopatologia , Animais , Progressão da Doença , Portadores de Fármacos/administração & dosagem , Resistência a Múltiplos Medicamentos/fisiologia , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Humanos , Receptores de Hialuronatos/administração & dosagem , Ácido Hialurônico/administração & dosagem , Ácido Hialurônico/fisiologia , Hialuronoglucosaminidase/uso terapêutico , Modelos Biológicos , Nanoconjugados/administração & dosagem , Neoplasias/tratamento farmacológico , Isoformas de Proteínas/administração & dosagem , Isoformas de Proteínas/fisiologia
7.
Haematologica ; 95(7): 1191-7, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20081055

RESUMO

BACKGROUND: Recently, we demonstrated immunological and clinical responses to a RHAMM-R3 peptide vaccine in patients with acute myeloid leukemia, myelodysplastic syndrome and multiple myeloma. To improve the outcome of the vaccine, a second cohort was vaccinated with a higher dose of 1,000 microg peptide. DESIGN AND METHODS: Nine patients received four vaccinations subcutaneously at a biweekly interval. Immunomonitoring of cytotoxic CD8(+) as well as regulatory CD4(+) T cells was performed by flow cytometry as well as by enzyme-linked immunospot (ELISpot) assays. Parameters of clinical response were assessed. RESULTS: In 4 of 9 patients (44%) we detected positive immunological responses. These patients showed an increase of CD8(+)RHAMM-R3_tetramer(+)/CD45RA(+)/CCR7(-)/CD27(-)/CD28(-) effector T cells and an increase of R3-specific CD8+ T cells. Two of these patients showed a significant decrease of regulatory T cells (Tregs). In one patient without response Tregs frequency increased from 5 to 16%. Three patients showed clinical effects: one patient with myelodysplastic syndrome RAEB-1 showed a reduction of leukemic blasts in the bone marrow, another myelodysplastic syndrome patient an improvement of peripheral blood counts and one patient with multiple myeloma a reduction of free light chains. Clinical and immunological reactions were lower in this cohort than in the 300 microg cohort. CONCLUSIONS: High-dose RHAMM-R3 peptide vaccination induced immunological responses and positive clinical effects. Therefore, RHAMM constitutes a promising structure for further targeted immunotherapies in patients with different hematologic malignancies. However, higher doses of peptide did not improve the frequency and intensity of immune responses in this trial.


Assuntos
Vacinas Anticâncer/administração & dosagem , Proteínas da Matriz Extracelular/administração & dosagem , Receptores de Hialuronatos/administração & dosagem , Imunidade/efeitos dos fármacos , Fragmentos de Peptídeos/administração & dosagem , Idoso , Vacinas Anticâncer/química , Relação Dose-Resposta a Droga , Proteínas da Matriz Extracelular/imunologia , Feminino , Humanos , Receptores de Hialuronatos/imunologia , Masculino , Pessoa de Meia-Idade , Fragmentos de Peptídeos/imunologia , Linfócitos T/imunologia , Vacinação
8.
J Immunol ; 181(7): 4676-84, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18802070

RESUMO

Cachexia is the dramatic weight loss and muscle atrophy seen in chronic disease states, including autoimmunity, cancer, and infection, and is often associated with lymphopenia. We have previously shown that CD4(+) T cells that express the lowest density of CD44 (CD4(+)CD44(v.low)) are significantly reduced in diabetic NOD mice that are cachexic compared with diabetic mice that are not cachexic. Using this model, and a model of cancer cachexia, we test the hypothesis that CD4(+)CD44(v.low) cells play an active role in protecting the host from cachexia. CD4(+)CD44(v.low) cells, but not CD4(+) cells depleted of CD44(v.low) cells, delay the onset of wasting when infused into either diabetic or prediabetic NOD recipients. However, no significant effect on the severity of diabetes was detected. In a model of cancer cachexia, they significantly reduce muscle atrophy, and inhibit muscle protein loss and DNA loss, even when given after the onset of cachexia. Protection from wasting and muscle atrophy by CD4(+)CD44(v.low) cells is associated with protection from lymphopenia. These data suggest, for the first time, a role for an immune cell subset in protection from cachexia, and further suggest that the mechanism of protection is independent of protection from autoimmunity.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/transplante , Caquexia/imunologia , Caquexia/terapia , Animais , Linfócitos T CD4-Positivos/metabolismo , Caquexia/prevenção & controle , Carcinoma Pulmonar de Lewis , Linhagem Celular Tumoral , Diabetes Mellitus Tipo 1/diagnóstico , Diabetes Mellitus Tipo 1/imunologia , Feminino , Receptores de Hialuronatos/administração & dosagem , Receptores de Hialuronatos/biossíntese , Incidência , Linfopenia/imunologia , Linfopenia/terapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Atrofia Muscular/imunologia , Atrofia Muscular/prevenção & controle , Transplante de Neoplasias , Índice de Gravidade de Doença , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/transplante , Síndrome de Emaciação/imunologia , Síndrome de Emaciação/terapia
9.
Biochem Biophys Res Commun ; 343(2): 609-16, 2006 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-16554035

RESUMO

Bone marrow stromal cells produce large amounts of extracellular matrix and cytokines. Amongst them, hyaluronan, a glycosaminoglycan and ligand for the cell surface molecule CD44, and TGFbeta1, a cytokine particularly important in monocyte differentiation. We have studied in vitro the role of hyaluronan and TGFbeta1 in the differentiation process of U937 monocytic progenitor cells. We provide evidence that, in the presence of whole blood-derived serum, the addition of hyaluronan is sufficient to induce the expression of NADPH-oxidase components but not of other monocytic markers (CD14, CD11b, and VLA-4). In the presence of plasma-derived serum, besides hyaluronan, the additional presence of TGFbeta1 was required for the expression of all of the components of the NADPH oxidase. We further show that hyaluronan mediates its effect through CD44. We conclude that cell matrix factors act cooperatively with cytokines to induce the expression of the components of the NADPH-oxidase in monocytic progenitor cells.


Assuntos
Células Precursoras de Granulócitos/citologia , Células Precursoras de Granulócitos/enzimologia , Receptores de Hialuronatos/administração & dosagem , Monócitos/citologia , Monócitos/enzimologia , NADPH Oxidases/metabolismo , Fator de Crescimento Transformador beta/administração & dosagem , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Sinergismo Farmacológico , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Células Precursoras de Granulócitos/efeitos dos fármacos , Humanos , Monócitos/efeitos dos fármacos , Fator de Crescimento Transformador beta1
10.
J Immunol ; 163(3): 1619-27, 1999 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-10415067

RESUMO

At sites of chronic inflammation seen during infections, autoimmunity, graft-vs-host response, and cytokine therapy, endothelial cell injury is known to occur, the exact mechanism of which is unknown. In the current study we used IL-2-induced vascular leak syndrome (VLS) as a model to investigate whether cytotoxic lymphocytes use CD44 in mediating endothelial cell injury. Administration of IL-2 to wild-type mice triggered significant VLS in the lungs and liver. In contrast, in CD44 knockout (KO) mice, IL-2-induced VLS was markedly reduced in the lungs and liver. IL-2-treated wild-type and CD44 KO mice had similar levels of perivascular infiltration with lymphocytes in the lungs and liver. This suggested that the decrease in VLS seen in CD44 KO mice was not due to the inability of lymphocytes to migrate to these organs. Ultrastructural studies demonstrated extensive endothelial cell damage in the lungs and liver of IL-2-treated wild-type, but not CD44 KO, mice. Moreover, CD44-KO mice exhibited a marked decrease in IL-2-induced lymphokine-activated killer cell activity. The induction of VLS was dependent on the expression of CD44 on immune cells rather than endothelial cells because adoptive transfer of CD44+, but not CD44- spleen cells along with IL-2 into CD44 KO mice triggered VLS. The IL-2-induced VLS was blocked by administration of F(ab')2 of Abs against CD44. The current study demonstrates that CD44 plays a key role in endothelial cell injury. Blocking CD44 in vivo may offer a novel therapeutic approach to prevent endothelial cell injury by cytotoxic lymphocytes in a variety of clinical disease models.


Assuntos
Síndrome de Vazamento Capilar/imunologia , Endotélio Vascular/imunologia , Endotélio Vascular/patologia , Receptores de Hialuronatos/imunologia , Transferência Adotiva , Animais , Anticorpos Monoclonais/administração & dosagem , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Síndrome de Vazamento Capilar/etiologia , Síndrome de Vazamento Capilar/genética , Síndrome de Vazamento Capilar/patologia , Células Cultivadas , Citotoxicidade Imunológica/genética , Endotélio Vascular/química , Feminino , Receptores de Hialuronatos/administração & dosagem , Receptores de Hialuronatos/biossíntese , Receptores de Hialuronatos/genética , Ácido Hialurônico/farmacologia , Fragmentos Fab das Imunoglobulinas/administração & dosagem , Injeções Intraperitoneais , Interleucina-2/administração & dosagem , Células Matadoras Ativadas por Linfocina/imunologia , Fígado/patologia , Fígado/ultraestrutura , Pulmão/patologia , Pulmão/ultraestrutura , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Baço/citologia , Baço/transplante , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA