Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
J Exp Med ; 190(3): 341-54, 1999 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-10430623

RESUMO

Neovascularization is essential for growth and spread of primary and metastatic tumors. We have identified a novel cytokine, endothelial-monocyte activating polypeptide (EMAP) II, that potently inhibits tumor growth, and appears to have antiangiogenic activity. Mice implanted with Matrigel showed an intense local angiogenic response, which EMAP II blocked by 76% (P < 0.001). Neovascularization of the mouse cornea was similarly prevented by EMAP II (P < 0.003). Intraperitoneally administered EMAP II suppressed the growth of primary Lewis lung carcinomas, with a reduction in tumor volume of 65% versus controls (P < 0.003). Tumors from human breast carcinoma-derived MDA-MB 468 cells were suppressed by >80% in EMAP II-treated animals (P < 0.005). In a lung metastasis model, EMAP II blocked outgrowth of Lewis lung carcinoma macrometastases; total surface metastases were diminished by 65%, and of the 35% metastases present, approximately 80% were inhibited with maximum diameter <2 mm (P < 0.002 vs. controls). In growing capillary endothelial cultures, EMAP II induced apoptosis in a time- and dose-dependent manner, whereas other cell types were unaffected. These data suggest that EMAP II is a tumor-suppressive mediator with antiangiogenic properties allowing it to target growing endothelium and limit establishment of neovasculature.


Assuntos
Apoptose , Citocinas , Endotélio Vascular/fisiologia , Inibidores do Crescimento/fisiologia , Proteínas de Neoplasias/fisiologia , Proteínas de Ligação a RNA/fisiologia , Células Tumorais Cultivadas/patologia , Animais , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Lewis , Bovinos , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/farmacologia , Inibidores do Crescimento/sangue , Inibidores do Crescimento/genética , Inibidores do Crescimento/farmacocinética , Humanos , Infusões Intravenosas , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas de Neoplasias/sangue , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/farmacocinética , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/genética , Proteínas de Ligação a RNA/sangue , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/farmacocinética , Proteínas Recombinantes/sangue , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacologia , Distribuição Tecidual/genética , Células Tumorais Cultivadas/efeitos dos fármacos
2.
Nutr Cancer ; 61(4): 538-43, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19838926

RESUMO

The effect of macelignan, a phytoestrogen, on P-gp function was investigated using multidrug resistant cancer cells overexpressing P-gp (NCI/ADR-RES) and the fluorescent P-gp substrates, daunorubicin and rhodamine 123. Macelignan (40 microM) increased the cellular accumulation of daunorubicin by approximately threefold in NCI/ADR-RES cells, whereas it did not alter the cellular accumulation of daunorubicin in MCF-7/sensitive cells. Similarly, the presence of macelignan also enhanced significantly (P < 0.05) the cellular accumulation of rhodamine 123 in a concentration-dependent manner in NCI/ADR-RES cells. Furthermore, cancer cells were more susceptible to the cytotoxicity of vinblastine, a P-gp substrate, in the presence of macelignan. Those results suggest that macelignan has inhibitory effects on P-gp mediated cellular efflux. However, P-gp activity did not affect the cellular accumulation of macelignan itself. Taken all together, macelignan was identified as a novel inhibitor of P-gp activity and may be a promising lead compound for the rational design of more efficacious drugs to reverse multidrug resistance in cancer.


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/antagonistas & inibidores , Antineoplásicos Fitogênicos/farmacologia , Resistencia a Medicamentos Antineoplásicos , Lignanas/farmacologia , Adenocarcinoma , Análise de Variância , Antineoplásicos Fitogênicos/farmacocinética , Transporte Biológico Ativo/efeitos dos fármacos , Neoplasias da Mama , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Daunorrubicina/farmacocinética , Relação Dose-Resposta a Droga , Desenho de Fármacos , Resistência a Múltiplos Medicamentos , Sinergismo Farmacológico , Feminino , Corantes Fluorescentes/farmacocinética , Inibidores do Crescimento/farmacocinética , Inibidores do Crescimento/farmacologia , Humanos , Lignanas/farmacocinética , Fitoestrógenos/farmacocinética , Fitoestrógenos/farmacologia , Fitoterapia , Rodamina 123/farmacocinética , Vimblastina/farmacocinética
3.
Molecules ; 13(7): 1441-54, 2008 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-18719516

RESUMO

A series of amino acid monoester prodrugs of floxuridine was synthesized and evaluated for the improvement of oral bioavailability and the feasibility of target drug delivery via oligopeptide transporters. All floxuridine 5'-amino acid monoester prodrugs exhibited PEPT1 affinity, with inhibition coefficients of Gly-Sar uptake (IC50) ranging from 0.7 - 2.3 mM in Caco-2 and 2.0 - 4.8 mM in AsPC-1 cells, while that of floxuridine was 7.3 mM and 6.3 mM, respectively. Caco-2 membrane permeabilities of floxuridine prodrugs (1.01 - 5.31 x 10(-6 )cm/sec) and floxuridine (0.48 x 10(-6 )cm/sec) were much higher than that of 5-FU (0.038 x 10(-6) cm/sec). MDCK cells stably transfected with the human oligopeptide transporter PEPT1 (MDCK/hPEPT1) exhibited enhanced cell growth inhibition in the presence of the prodrugs. This prodrug strategy offers great potential, not only for increased drug absorption but also for improved tumor selectivity and drug efficacy.


Assuntos
Aminoácidos/farmacologia , Antimetabólitos Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Floxuridina/farmacologia , Floxuridina/farmacocinética , Pró-Fármacos/farmacologia , Pró-Fármacos/farmacocinética , Simportadores/metabolismo , Absorção/efeitos dos fármacos , Aminoácidos/síntese química , Aminoácidos/farmacocinética , Animais , Antimetabólitos Antineoplásicos/síntese química , Células CACO-2 , Cães , Sistemas de Liberação de Medicamentos , Ésteres , Inibidores do Crescimento/farmacocinética , Inibidores do Crescimento/farmacologia , Humanos , Transportador 1 de Peptídeos , Pró-Fármacos/síntese química , Simportadores/biossíntese , Simportadores/genética
4.
Sci Rep ; 7: 40142, 2017 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-28071663

RESUMO

Prosthetic grafts and patches are commonly used in cardiovascular surgery, however neointimal hyperplasia remains a significant concern, especially under low flow conditions. We hypothesized that delivery of rapamycin from nanoparticles (NP) covalently attached to patches allows sustained site-specific delivery of therapeutic agents targeted to inhibit localized neointimal hyperplasia. NP were covalently linked to pericardial patches using EDC/NHS chemistry and could deliver at least 360 ng rapamycin per patch without detectable rapamycin in serum; nanoparticles were detectable in the liver, kidney and spleen but no other sites within 24 hours. In a rat venous patch angioplasty model, control patches developed robust neointimal hyperplasia on the patch luminal surface characterized by Eph-B4-positive endothelium and underlying SMC and infiltrating cells such as macrophages and leukocytes. Patches delivering rapamycin developed less neointimal hyperplasia, less smooth muscle cell proliferation, and had fewer infiltrating cells but retained endothelialization. NP covalently linked to pericardial patches are a novel composite delivery system that allows sustained site-specific delivery of therapeutics; NP delivering rapamycin inhibit patch neointimal hyperplasia. NP linked to patches may represent a next generation of tissue engineered cardiovascular implants.


Assuntos
Angioplastia/métodos , Inibidores do Crescimento/administração & dosagem , Hiperplasia/prevenção & controle , Nanopartículas/administração & dosagem , Neointima/patologia , Sirolimo/administração & dosagem , Transplantes/cirurgia , Animais , Portadores de Fármacos/administração & dosagem , Inibidores do Crescimento/farmacocinética , Histocitoquímica , Hiperplasia/patologia , Ratos , Sirolimo/farmacocinética , Resultado do Tratamento
5.
Eur J Pharm Sci ; 97: 70-78, 2017 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-27816627

RESUMO

The purpose of this study was to establish a population pharmacokinetic/pharmacodynamic (PK/PD) model linking etoposide free tumor and total plasma concentrations to the inhibition of solid tumor growth in rats. Walker-256 tumor cells were inoculated subcutaneously in the right flank of Wistar rats, which were randomly divided in control and two treated groups that received etoposide 5 or 10mg/kg i.v. bolus every day for 8 and 4days, respectively, and tumor volume was monitored daily for 30days. The plasma and intratumoral concentrations-time profiles were obtained from a previous study and were modeled by a four-compartment population pharmacokinetic (popPK) model. PK/PD analysis was conducted using MONOLIX v.4.3.3 on average data and by mean of a nonlinear mixed-effect model. PK/PD data were analyzed using a modification of Simeoni Tumor Growth Inhibition (TGI) model by introduction of an Emax function to take into account the concentration dependency of k2variable parameter (variable potency). The Simeoni TGI-Emax model was capable to fit schedule-dependent antitumor effects using the tumor growth curves from the control and two different administered schedules. The PK/PD model was capable of describing the tumor growth inhibition using total plasma or free tumor concentrations, resulting in higher k2max (maximal potency) for free concentrations (25.8mL·µg-1·day-1 - intratumoral vs. 12.6mL·µg-1·day-1 total plasma). These findings indicate that the plasma concentration may not be a good surrogate for pharmacologically active free tumor concentrations, emphasizing the importance of knowing drug tumor penetration to choose the best antitumor therapy.


Assuntos
Antineoplásicos Fitogênicos/farmacocinética , Carcinoma 256 de Walker/metabolismo , Modelos Animais de Doenças , Etoposídeo/farmacocinética , Inibidores do Crescimento/farmacocinética , Carga Tumoral/efeitos dos fármacos , Animais , Antineoplásicos Fitogênicos/uso terapêutico , Carcinoma 256 de Walker/tratamento farmacológico , Carcinoma 256 de Walker/patologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Etoposídeo/uso terapêutico , Inibidores do Crescimento/uso terapêutico , Masculino , Ratos , Ratos Wistar , Carga Tumoral/fisiologia
6.
Cancer Res ; 61(5): 1983-90, 2001 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-11280756

RESUMO

4-demethoxy-3'-deamino-3'-aziridinyl-4'-methylsulphonyl-daunorubicin (PNU-159548) belongs to a novel class of antitumor compounds (termed alkycyclines) and is currently undergoing Phase II clinical trial. In the present study, we investigated the in vitro and in vivo antitumor activity, the pharmacokinetics, and the toxicological profile of this compound. PNU-159548 showed good cytotoxic activity in murine and human cancer cells growing in vitro, with an average concentration for 50% growth inhibition of 15.8 ng/ml. The drug showed strong antitumor efficacy in vivo after i.v. and p.o. administration against rapidly proliferating murine leukemias and slowly growing transplantable human xenografts. At non-toxic doses, PNU-159548 produced complete regression and cures in ovarian, breast, and human small cell lung carcinomas. Fourteen of 16 models studied, including colon, pancreatic, gastric, and renal carcinomas, astrocytoma and melanoma, were found to be sensitive to PNU-159548. In addition, PNU-159548 was effective against intracranially implanted tumors. Toxicological studies revealed myelosuppression as the main toxicity in both mice and dogs. The maximum tolerated doses, after a single administration, were 2.5 mg/kg of body weight in mice, 1.6 mg/kg in rats, and 0.3 mg/kg in dogs. In the cyclic studies, the maximum tolerated doses were 0.18 mg/kg/day (cumulative dose/cycle: 0.54 mg/kg) in rats and 0.05 mg/kg/day (cumulative dose/cycle: 0.15 mg/kg) in dogs. PNU-159548 showed minimal cardiotoxicity, when compared with doxorubicin in the chronic rat model at a dose level inducing similar myelotoxicity. Animal pharmacokinetics, carried out in mice, rats, and dogs, was characterized by high volumes of distribution, plasma clearance of the same order of the hepatic blood flow, and short terminal half-life. These findings support the conclusion that PNU-159548 is an excellent candidate for clinical trials in the treatment of cancer.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Daunorrubicina/farmacologia , Animais , Antibióticos Antineoplásicos/farmacocinética , Antibióticos Antineoplásicos/toxicidade , Cardiomiopatias/induzido quimicamente , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Daunorrubicina/análogos & derivados , Daunorrubicina/farmacocinética , Daunorrubicina/toxicidade , Ensaios de Seleção de Medicamentos Antitumorais , Inibidores Enzimáticos/farmacocinética , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/toxicidade , Feminino , Inibidores do Crescimento/farmacocinética , Inibidores do Crescimento/farmacologia , Inibidores do Crescimento/toxicidade , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos DBA , Inibidores da Topoisomerase II , Células Tumorais Cultivadas/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Cancer Res ; 61(9): 3669-74, 2001 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-11325837

RESUMO

The therapeutic efficacy and tumor accumulation of a liposome formulation of 1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea (CCNU), an effective agent used in the treatment of malignant brain tumors, was examined in an animal tumor model. Pharmacokinetic studies in normal and tumor-bearing rats indicated that a 2-fold greater plasma exposure was achieved with liposome-formulated CCNU compared with the free drug. In Fisher rats bearing s.c. tumors 36B-10, tumor growth was delayed substantially when liposomal CCNU was delivered compared with free-drug treatment. In single-dose treatments of 20, 35, and 50 mg/kg, tumor progression after each dose was reduced approximately 2-fold with liposomal compared with free CCNU (four animals in each treatment group). Multiple-dose treatments (given as three weekly doses with eight animals in each treatment group) with cumulative doses of 80 and 100 mg/kg of free and liposomal CCNU also resulted in a 2-fold reduction in tumor progression when compared with free-drug treatment. When drug levels in tumors relative to plasma were examined, it was observed that tumor drug concentrations did not exceed those found in plasma after administration of free CCNU; after administration of liposomal CCNU, however, tumor concentrations exceeded those in plasma by nearly 10-fold. These results suggest that the increased efficacy of liposome-formulated CCNU may be attributable to enhanced drug accumulation in tumor tissues.


Assuntos
Antineoplásicos Alquilantes/administração & dosagem , Astrocitoma/tratamento farmacológico , Neoplasias Encefálicas/tratamento farmacológico , Lomustina/administração & dosagem , Animais , Antineoplásicos Alquilantes/sangue , Antineoplásicos Alquilantes/farmacocinética , Antineoplásicos Alquilantes/farmacologia , Astrocitoma/sangue , Astrocitoma/metabolismo , Neoplasias Encefálicas/sangue , Neoplasias Encefálicas/metabolismo , Divisão Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Inibidores do Crescimento/administração & dosagem , Inibidores do Crescimento/sangue , Inibidores do Crescimento/farmacocinética , Inibidores do Crescimento/farmacologia , Lipossomos , Lomustina/sangue , Lomustina/farmacocinética , Lomustina/farmacologia , Ratos , Ratos Endogâmicos F344
8.
J Clin Oncol ; 17(4): 1095-104, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10561166

RESUMO

PURPOSE: To evaluate the clinical feasibility and pharmacologic behavior of the platelet-derived growth factor (PDGF) tyrosine kinase inhibitor SU101, administered on a prolonged, intermittent dosing schedule to patients with advanced solid malignancies. PATIENTS AND METHODS: Twenty-six patients were treated with SU101 doses ranging from 15 to 443 mg/m(2) as a 24-hour continuous intravenous (IV) infusion weekly for 4 weeks, repeated every 6 weeks. Pharmacokinetic studies were performed to characterize the disposition of SU101 and its major active metabolite, SU0020. Immunohistochemical staining of PDGF-alpha and -beta receptors was performed on malignant tumor specimens obtained at diagnosis. RESULTS: Twenty-six patients were treated with 52 courses (187 infusions) of SU101. The most common toxicities were mild to moderate nausea, vomiting, and fever. Two patients experienced one episode each of grade 3 neutropenia at the 333 and 443 mg/m(2) dose levels. Dose escalation of SU101 above 443 mg/m(2)/wk was precluded by the total volume of infusate required, 2.5 to 3.0 L. Individual plasma SU101 and SU0020 concentrations were described by a one-compartment model that incorporates both first-order formation and elimination of SU0020. SU101 was rapidly converted to SU0020, which exhibited a long elimination half-life averaging 19 +/- 12 days. At the 443 mg/m(2)/wk dose level, trough plasma SU0020 concentrations during weeks 2 and 4 ranged from 54 to 522 micromol/L. Immunohistochemical studies revealed PDGF-alpha and -beta receptor staining in the majority (15 of 19) of malignant neoplasms. CONCLUSION: SU101 was well tolerated as a 24-hour continuous IV infusion at doses of up to 443 mg/m(2)/wk for 4 consecutive weeks every 6 weeks. Although further dose escalation was precluded by infusate volume constraints, this SU101 dose schedule resulted in the achievement and maintenance of substantial plasma concentrations of the major metabolite, SU0020, for the entire treatment period.


Assuntos
Inibidores do Crescimento/farmacocinética , Inibidores do Crescimento/uso terapêutico , Isoxazóis/farmacocinética , Isoxazóis/uso terapêutico , Neoplasias/tratamento farmacológico , Adulto , Idoso , Cromatografia Líquida de Alta Pressão , Feminino , Inibidores do Crescimento/administração & dosagem , Humanos , Imuno-Histoquímica , Infusões Intravenosas , Isoxazóis/administração & dosagem , Leflunomida , Masculino , Pessoa de Meia-Idade , Neoplasias/metabolismo , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores do Fator de Crescimento Derivado de Plaquetas/análise , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
9.
Clin Cancer Res ; 5(12): 3977-82, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10632328

RESUMO

Abnormally regulated signaling through proliferative signal transduction pathways characterizes many of the common solid tumors. The best described of these involves potentially oncogenic proteins of the Ras family, which activate Raf proteins in the early steps of the mitogen-activated protein kinase cascade. ISIS 5132, a phosphorothioate antisense oligodexoynucleotide directed to the 3' untranslated region of the c-raf-1 mRNA, inhibits the growth of human tumor cell lines in vitro and in vivo in association with specific down-regulation of target message expression. Using a semiquantitative reverse transcription-PCR assay, we analyzed changes in c-raf-1 mRNA expression in peripheral blood mononuclear cells collected from patients with advanced cancers treated with ISIS 5132 as part of a clinical trial. Specimens were collected for analysis pretreatment and on days 3, 5, 8, and 15 of the first cycle and on day 1 of each subsequent cycle. We observed significant reductions of c-raf-1 expression from baseline by day 3 in 13 of 14 patients (P = 0.002). The time course and depletion of c-raf-1 message in peripheral blood mononuclear cells paralleled the clinical benefit in two patients. These findings demonstrate that ISIS 5132 specifically reduces target gene expression in treated patients and that peripheral blood mononuclear cells are suitable tissues for biomarker studies in future trials.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Oligodesoxirribonucleotídeos Antissenso/uso terapêutico , Proteínas Proto-Oncogênicas c-raf/antagonistas & inibidores , Tionucleotídeos/uso terapêutico , Idoso , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Relação Dose-Resposta a Droga , Esquema de Medicação , Feminino , Inibidores do Crescimento/efeitos adversos , Inibidores do Crescimento/farmacocinética , Inibidores do Crescimento/uso terapêutico , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias/enzimologia , Neoplasias/patologia , Oligodesoxirribonucleotídeos Antissenso/efeitos adversos , Oligodesoxirribonucleotídeos Antissenso/farmacocinética , Proteínas Proto-Oncogênicas c-raf/biossíntese , Proteínas Proto-Oncogênicas c-raf/genética , RNA Mensageiro/antagonistas & inibidores , Tionucleotídeos/efeitos adversos , Tionucleotídeos/farmacocinética
10.
Clin Cancer Res ; 4(6): 1557-61, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9626477

RESUMO

The N',N"-bis(ethyl) polyamine analogues demonstrate great potential as chemotherapeutic agents for lung cancer. This study examines how the expression of two oncogenes frequently associated with a worsened prognosis in lung cancer, c-myc and mutated ras, as well as the phenotypic transition induced by these genes, affects the sensitivity of small cell lung cancer (SCLC) cells to these polyamine analogues. Treatment with N1,N12-bis(ethyl)spermine (BE-Spm), a representative analogue, depresses polyamine levels and is cytostatic for the NCI H209 classic SCLC cell line. Both the overexpression of c-myc and the expression of oncogenic v-Ha-ras in these cells produce phenotypes that retain sensitivity to this growth inhibition. This sensitivity to BESpm is mediated by distinct pathways in these oncogene-expressing cells. c-myc overexpression markedly increases the expression of ornithine decarboxylase, which is then down-regulated by BESpm. In contrast, v-Ha-ras expression highly induces the polyamine catabolic enzyme spermidine/spermine N1-acetyltransferase. These findings suggest that the bis(ethyl)polyamine compounds may have broad utility for the treatment of both SCLC and non-SCLC, including those expressing oncogenic c-myc and ras.


Assuntos
Antineoplásicos/toxicidade , Genes myc , Genes ras , Inibidores do Crescimento/toxicidade , Poliaminas/metabolismo , Espermina/análogos & derivados , Acetiltransferases/metabolismo , Antineoplásicos/farmacocinética , Carcinoma de Células Pequenas , Divisão Celular/efeitos dos fármacos , Inibidores do Crescimento/farmacocinética , Humanos , Cinética , Neoplasias Pulmonares , Proteína Oncogênica p21(ras)/biossíntese , Proteínas Proto-Oncogênicas c-myc/biossíntese , Espermina/farmacocinética , Espermina/toxicidade , Fatores de Tempo , Células Tumorais Cultivadas
11.
J Neuroimmunol ; 106(1-2): 172-80, 2000 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-10814795

RESUMO

Leukemia inhibitory factor (LIF) is a neurotrophic cytokine now under clinical investigation for its effects on the CNS. We studied its passage across the blood-brain barrier (BBB) from blood to brain and spinal cord. Although a large amount of LIF was reversibly associated with the cerebral vasculature, intact LIF did reach brain parenchyma. Multiple-time regression analysis showed ready access of LIF to the CNS at a rate much faster than that of the vascular marker albumin. Excess LIF inhibited the entry of 125I-LIF after administration i.v. or by in-situ perfusion in blood-free buffer. Efflux of LIF from brain to blood was slower than reabsorption by CSF bulk flow, indicating that LIF tended to be retained in the brain. Although ciliary neurotrophic factor (CNTF) and LIF bind to the same receptor complex, CNTF did not cross-inhibit the entry of LIF into the CNS. A monoclonal antibody to LIF, however, abolished the entry of LIF. Our results show that peripherally administered LIF readily enters the brain and spinal cord by a saturable transport system across the BBB that may have biological implications.


Assuntos
Sistema Nervoso Central/metabolismo , Inibidores do Crescimento/farmacocinética , Interleucina-6 , Linfocinas/farmacocinética , Animais , Anticorpos/farmacologia , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/fisiologia , Fator Neurotrófico Ciliar/farmacologia , Relação Dose-Resposta a Droga , Estabilidade de Medicamentos , Inibidores do Crescimento/antagonistas & inibidores , Inibidores do Crescimento/sangue , Inibidores do Crescimento/imunologia , Humanos , Fator Inibidor de Leucemia , Linfocinas/antagonistas & inibidores , Linfocinas/sangue , Linfocinas/imunologia , Camundongos , Proteínas Recombinantes/sangue , Proteínas Recombinantes/farmacocinética , Distribuição Tecidual
13.
J Control Release ; 94(1): 63-74, 2004 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-14684272

RESUMO

Systemic application of anticancer drugs often causes severe toxic side effects. To reduce the undesired effects, advanced drug delivery systems are needed which are based on specific cell targeting vehicles. In this study, bacterial ghosts from Mannheimia haemolytica were used for site-specific delivery of doxorubicin (DOX) to human colorectal adenocarcinoma cells (Caco-2). Bacterial ghosts are non-denatured envelopes of Gram-negative bacteria with fully intact surface structures for specific attachment to mammalian cells. The in vitro release profile of DOX-ghosts demonstrated that the loaded drug was non-covalently associated with the bacterial ghosts and that the drug delivery vehicles themselves represent a slow release system. Adherence studies showed that the M. haemolytica ghosts more efficiently than E. coli ghosts targeted the Caco-2 cells and released the loaded DOX within the cells. Cytotoxicity assays revealed that the DOX-ghosts exhibited potent antiproliferative activities on Caco-2 cells as the DOX associated with ghosts was two magnitude of orders more cytotoxic than free DOX provided in the medium at the same concentrations. Notably, a significant reduction in the cell viability was measured with DOX-ghosts at low DOX concentrations, which had no inhibitory effect when applied as free DOX after incubation for 16 h or when applied at higher concentrations for only 10 min to the cells. As the higher antiproliferative effects of DOX on Caco-2 cells were mediated by the specific drug targeting properties of the bacterial ghosts, the bacterial ghost system represents a novel platform for advanced drug delivery.


Assuntos
Doxorrubicina/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Inibidores do Crescimento/administração & dosagem , Mannheimia haemolytica , Células CACO-2 , Relação Dose-Resposta a Droga , Doxorrubicina/farmacocinética , Inibidores do Crescimento/farmacocinética , Humanos , Mannheimia haemolytica/metabolismo
14.
Neurosci Lett ; 205(3): 202-6, 1996 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-8852593

RESUMO

Macrophage inflammatory proteins (MIP)-1 alpha and -1 beta have been postulated to exert their pyrogenic effects by acting directly at sites within the brain. Such activity would require circulating MIP-1s to cross the blood-brain barrier (BBB). We examined the ability of the monomer and polymer of MIP-1 alpha and the polymer of MIP-1 beta radioactively labeled with 125iodine (I-MIP-1) to cross the BBB. These I-MIP-1s behaved very similarly to each other but in a manner not previously seen for other cytokines. The I-MIP-1s immediately associated to a high degree and in a reversible manner with the vascular space of the brain. This association did not increase over time nor was it self-inhibitable. These results make it unlikely that the MIP-1s are transported into the brain by saturable transport systems in the manner found for some of the other cytokines. Other mechanisms, such as interactions with brain endothelia, leakage into brain through extracellular pathways, and binding at circumventricular organs, are more likely to provide the mechanisms through which blood-borne MIP-1s affect the central nervous system.


Assuntos
Barreira Hematoencefálica/fisiologia , Inibidores do Crescimento/farmacocinética , Proteínas Inflamatórias de Macrófagos/farmacocinética , Animais , Transporte Biológico/fisiologia , Quimiocina CCL4 , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Injeções Intravenosas , Masculino , Camundongos , Camundongos Endogâmicos ICR , Proteínas Recombinantes/farmacocinética , Análise de Regressão
15.
Anticancer Res ; 20(5B): 3591-601, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11131667

RESUMO

The relationship between growth and the antioxidant enzyme defence system in human MCF-7 (breast) cancer cells treated with bovine milk fat enriched with conjugated linoleic acid (CLA) was studied. Milk enriched in CLA was obtained from cows on pasture supplemented with full fat rapeseeds and full fat soyabeans (1). Cell number decreased up to 90% (p < 0.05) and lipid peroxidation increased 15-fold (p < 0.05) following incubation of MCF-7 cells for 8 days with increasing levels of milk fat yielding CLA concentrations between 16.9 and 22.6 ppm. Growth suppression and prooxidant effects of milk fat CLA were independent of the variable composition of the milk fat samples, suggesting that CLA was the active ingredient in milk fat responsible for the cytotoxic effect. Mixtures containing isomers of CLA (c9, t11-, t10, c12-, c11, t13- and minor amounts of other isomers) and linoleic acid (LA) at similar concentrations to the milk fat samples were as effective at inhibiting growth and stimulating peroxidation of MCF-7 cells as the milk fatty acids. Incubation of the cells with the c9, t11 CLA isomer (20 ppm) or the mixture of CLA isomers (20 ppm) for 8 days resulted in a 60% decrease (p < 0.05) in viability compared with untreated controls and was significantly (p < 0.05) more effective than incubation with the t10, c12 CLA isomer (20 ppm), which caused only a 15% decrease in cell numbers under similar conditions. A 25% increase (p < 0.05) in cell proliferation occurred when LA (20 ppm) alone was incubated with MCF-7 cells for 8 days. 14C-CLA was preferentially incorporated into the phospholipid fraction of the MCF-7 cell lipids in a dose-dependent manner and CLA accumulated in cell membranes more efficiently when the cells were incubated in the presence of milk fat than the c9, t11 synthetic CLA isomer. Superoxide dismutase (SOD), catalase and glutathione peroxidase (GPx) activities were induced in MCF-7 cells exposed to milk fat (containing 16.9-22.6 ppm CLA) over 8 days. The data indicate that milk fat triglyceride-bound CLA, consisting primarily of the c9, t11 isomer, was cytotoxic towards MCF-7 cells.


Assuntos
Neoplasias da Mama/patologia , Ácidos Graxos/farmacologia , Inibidores do Crescimento/farmacologia , Ácidos Linoleicos/farmacologia , Leite/química , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Radioisótopos de Carbono , Catalase/metabolismo , Bovinos , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Dieta , Relação Dose-Resposta a Droga , Ácidos Graxos/farmacocinética , Feminino , Glutationa Peroxidase/metabolismo , Inibidores do Crescimento/farmacocinética , Humanos , Ácidos Linoleicos/farmacocinética , Peroxidação de Lipídeos/efeitos dos fármacos , Glycine max , Superóxido Dismutase/metabolismo , Células Tumorais Cultivadas
16.
Drug Deliv ; 10(2): 95-100, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12746055

RESUMO

Some selected lipophilic conjugates of the antifolate drug methotrexate (MTX) with lipoamino acids (LAA), previously described, were incorporated in liposomes with a different composition and charge (neutral, positive, or negative). The properties of the liposomal systems were determined. The inhibitory activity of the conjugates after incorporation in the vesicles was determined in a preliminary assessment against a human erythroleukemic cell line (K562 cells) and compared with the activity of the parent drug and of free conjugates. The influence of liposome surface charge and of the type of conjugate (i.e., in the carboxylic or ester form) on the biological effect is discussed.


Assuntos
Aminoácidos Neutros/administração & dosagem , Aminoácidos Neutros/farmacocinética , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Preparações de Ação Retardada/uso terapêutico , Portadores de Fármacos/farmacocinética , Metotrexato/administração & dosagem , Metotrexato/farmacocinética , Preparações de Ação Retardada/administração & dosagem , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/química , Combinação de Medicamentos , Avaliação Pré-Clínica de Medicamentos/métodos , Estabilidade de Medicamentos , Inibidores do Crescimento/administração & dosagem , Inibidores do Crescimento/farmacocinética , Humanos , Células K562 , Lipídeos/química , Lipossomos , Fosfolipídeos/química , Tecnologia Farmacêutica
17.
Biol Trace Elem Res ; 78(1-3): 241-54, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11314982

RESUMO

The metabolism of arsenic, its affinity to metallothionein (MT), its influence on selenium levels, and its biotransformation to different metabolites in the liver tissue of laying hens exposed to arsenic trioxide (As2O3) was investigated. The experiment was performed with two groups of hens fed for 19 d with either a standard diet or with the same diet enriched in arsenic (30 microg/g). The major findings were as follows: 1. After 19 d exposure, about 65% of the total liver As was found in the water-soluble phase (100,000g centrifuged supernatant). In liver supernatant, As binding was found mostly in the range of very low-molecular-weight proteins (Mr < 10,000). Although after exposure the amount of MT-like proteins increased, the As bound to it was only in trace amounts. The protein was identified by convential procedures as Zn,Cu-thionein with traces of selenium and arsenic. 2. Arsenic exposure resulted in almost unchanged Se levels regarding its tissue concentrations and distribution between supernatant and pellet, where about 10% of total Se was found in the supernatant. On the contrary, As exposure did affect Cd levels. Tissue Cd concentration was slightly diminished, but the percentage of tissue Cd found in the water-soluble phase was increased from 20% to 40%. 3. In methanol extracts of tissue and supernatant of the As-exposed group, only two arsenic compounds were detected, As(III) and dimethylarsinic acid (DMA), the latter prevailing.


Assuntos
Arsenicais/farmacologia , Galinhas , Inibidores do Crescimento/farmacologia , Fígado/metabolismo , Metalotioneína/metabolismo , Óxidos/farmacologia , Animais , Arsênio/metabolismo , Trióxido de Arsênio , Arsenicais/farmacocinética , Bioquímica/métodos , Cádmio/metabolismo , Cobre/metabolismo , Feminino , Inibidores do Crescimento/farmacocinética , Inativação Metabólica , Fígado/efeitos dos fármacos , Metalotioneína/efeitos dos fármacos , Metanol/química , Óxidos/farmacocinética , Selênio/metabolismo , Extratos de Tecidos/química , Zinco/metabolismo
18.
J Interferon Cytokine Res ; 34(10): 759-68, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24841172

RESUMO

Interferon gamma (IFN-γ) is a 28 kDa homodimeric cytokine that exhibits potent immunomodulatory, anti-proliferative, and antiviral properties. The protein is used to treat chronic granulomatous disease and malignant osteopetrosis, and it is under investigation as a treatment for a variety of cancer, fungal and viral diseases. IFN-γ has a short circulating half life in vivo, which necessitates frequent administration to patients. An unusual feature of IFN-γ is that the protein contains no native cysteines. To create a longer-acting and potentially more effective form of the protein, we introduced a cysteine residue into the IFN-γ coding sequence at amino acid position 103, which is located in a surface-exposed, non-helical region of the protein. The added cysteine residue served as the site for targeted modification of the protein with a cysteine-reactive polyethylene glycol (PEG) reagent. The recombinant protein was expressed in bacteria, purified and modified with 10, 20, and 40 kDa maleimide PEGs. The purified, PEGylated proteins had in vitro bioactivities comparable to IFN-γ, as measured using an in vitro cell growth inhibition assay. The PEGylated proteins displayed 20- to 32-fold longer half lives than IFN-γ in rats, and they were significantly more effective than IFN-γ at inhibiting growth of a human tumor xenograft in athymic mice.


Assuntos
Cisteína/química , Inibidores do Crescimento/farmacocinética , Interferon gama/farmacocinética , Polietilenoglicóis/química , Animais , Linhagem Celular Tumoral , Cisteína/genética , Feminino , Inibidores do Crescimento/química , Inibidores do Crescimento/farmacologia , Humanos , Imunomodulação , Interferon gama/química , Interferon gama/farmacologia , Masculino , Camundongos , Camundongos Nus , Mutação/genética , Ligação Proteica , Conformação Proteica , Engenharia de Proteínas , Estabilidade Proteica , Ratos , Ratos Sprague-Dawley , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Biochem Pharmacol ; 83(6): 715-22, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22222427

RESUMO

Hydrogen sulfide-releasing non-steroidal anti-inflammatory drugs (HS-NSAIDs) are an emerging novel class of compounds with significant anti-inflammatory properties. They consist of a traditional NSAID to which an H(2)S-releasing moiety is covalently attached. We examined the effects of four different HS-NSAIDs on the growth properties of eleven different human cancer cell lines of six different tissue origins. Human colon, breast, pancreatic, prostate, lung, and leukemia cancer cell lines were treated with HS-aspirin, -sulindac, -iburofen, -naproxen, and their traditional counterparts. HS-NSAIDs inhibited the growth of all cancer cell lines studied, with potencies of 28- to >3000-fold greater than that of their traditional counterparts. HS-aspirin (HS-ASA) was consistently the most potent. HS-NSAIDs inhibited cell proliferation, induced apoptosis, and caused G(0)/G(1) cell cycle block. Metabolism of HS-ASA by colon cells showed that the acetyl group of ASA was hydrolyzed rapidly, followed by hydrolysis of the ester bond linking the salicylate anion to the H(2)S releasing moiety, producing salicylic acid and ADT-OH from which H(2)S is released. In reconstitution studies, ASA and ADT-OH were individually less active than the intact HS-ASA towards cell growth inhibition. Additionally, the combination of these two components representing a fairly close approximation to the intact HS-ASA, was 95-fold less active than the intact HS-ASA for growth inhibition. Taken together, these results demonstrate that HS-NSAIDs have potential anti-growth activity against a wide variety of human cancer cells.


Assuntos
Anti-Inflamatórios não Esteroides/farmacocinética , Antineoplásicos/farmacocinética , Aspirina/análogos & derivados , Proliferação de Células/efeitos dos fármacos , Inibidores do Crescimento/farmacocinética , Sulfeto de Hidrogênio/metabolismo , Ibuprofeno/análogos & derivados , Naproxeno/análogos & derivados , Sulindaco/análogos & derivados , Tionas/farmacocinética , Anti-Inflamatórios não Esteroides/química , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Aspirina/química , Aspirina/farmacocinética , Linhagem Celular Tumoral , Inibidores do Crescimento/química , Humanos , Sulfeto de Hidrogênio/química , Ibuprofeno/química , Ibuprofeno/farmacocinética , Naproxeno/química , Naproxeno/farmacocinética , Sulindaco/química , Sulindaco/farmacocinética , Tionas/química
20.
Cancer Chemother Pharmacol ; 64(2): 425-9, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19280191

RESUMO

PURPOSE: The toxicities, pharmacokinetics and recommended dose of oral once daily ZK 304709, a novel multi-targeted growth inhibitor (MTGI) with activity against cell-cycle progression and angiogenesis, was investigated in patients by administration for 14 consecutive days followed by 14 days recovery. METHODS: Patients with solid tumours resistant to standard treatments were enrolled in an accelerated titration design. RESULTS: Thirty-seven patients received ZK 304709 from 15 to 285 mg daily. The most common drug-related adverse events were vomiting, diarrhoea and fatigue. Systemic exposure to ZK 304709 increased with dose up to 90 mg daily but plateaued thereafter, with high inter-individual variability at all doses. Thirteen patients had stable disease as best response as per RECIST criteria. CONCLUSIONS: There was no increase in exposure to ZK 304709 with dose escalation above 90 mg, and the MTD was not determined. This study illustrates the importance of phase I pharmacokinetic data to guide dose escalation and drug development.


Assuntos
Inibidores da Angiogênese/farmacocinética , Antineoplásicos/farmacocinética , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Inibidores do Crescimento/farmacocinética , Neoplasias/metabolismo , Administração Oral , Adulto , Idoso , Inibidores da Angiogênese/efeitos adversos , Antineoplásicos/efeitos adversos , Relação Dose-Resposta a Droga , Feminino , Inibidores do Crescimento/efeitos adversos , Humanos , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias/patologia , Prognóstico , Segurança , Taxa de Sobrevida , Distribuição Tecidual , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa