Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 202
Filtrar
1.
Reprod Toxicol ; 113: 71-84, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35961530

RESUMO

Melphalan significantly contributes to the increase in childhood cancer survival rate. It acts as a gonadotoxic agent and leads to testes damage, dysbalance in gonadal hormones, and impairment in the germ cell proliferation. Therefore, it might be a potent threat to male fertility in individuals who have undergone melphalan treatment during childhood cancer. However, the molecular mechanisms of melphalan-induced gonadal damage are not yet fully explored and they need to be investigated to determine the benefit-risk profile. In the present study, juvenile male SD rats were subjected to single and intermittent cycles of melphalan exposure in a dose-dependent (0.375, 0.75 and 1.5 mg/kg) manner. Methods of end-points evaluations were quantification of micronuclei formation in peripheral blood, sperm count, sperm motility and head morphology, sperm and testicular DNA damage, histological studies in testes, oxidative/nitrosative stress parameters. A single cycle of exposure at high dose (1.5 mg/kg) produced significant effect on micronuclei formation only after the first week of exposure, whereas failed to produce significant effect at the end of the sixth week. Intermittent cycles of exposure at the dose of 1.5 mg/kg produced significant alterations in all the parameters (micronuclei in peripheral blood, testes and epididymides weight and length, MDA, GSH and nitrite levels, sperm count and motility, sperm head morphology, testicular and sperm DNA damage, protein expression in testes and histological parameters). So, time of exposure as well as the amount of exposure (total dosage administered) is critical in determining the magnitude of the damage in germ cell risk assessment.


Assuntos
Melfalan , Motilidade dos Espermatozoides , Animais , Células Germinativas/metabolismo , Masculino , Melfalan/metabolismo , Melfalan/toxicidade , Nitritos/metabolismo , Estresse Oxidativo , Ratos , Ratos Sprague-Dawley , Medição de Risco , Sêmen/metabolismo , Contagem de Espermatozoides , Espermatozoides , Testículo
2.
Stem Cell Res Ther ; 11(1): 470, 2020 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-33153480

RESUMO

BACKGROUND: Treatment-induced cardiotoxicity is a leading noncancer-related cause of acute and late onset morbidity and mortality in cancer patients on antineoplastic drugs such as melphalan-increasing clinical case reports have documented that it could induce cardiotoxicity including severe arrhythmias and heart failure. As the mechanism by which melphalan impairs cardiac cells remains poorly understood, here, we aimed to use cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs) to investigate the cellular and molecular mechanisms of melphalan-induced cardiotoxicity. METHODS: hiPSC-CMs were generated and treated with clinically relevant doses of melphalan. To characterize melphalan-induced cardiotoxicity, cell viability and apoptosis were quantified at various treatment durations. Ca2+ transient and contractility analyses were used to examine the alterations of hiPSC-CM function. Proteomic analysis, reactive oxygen species detection, and RNA-Sequencing were conducted to investigate underlying mechanisms. RESULTS: Melphalan treatment of hiPSC-CMs induced oxidative stress, caused Ca2+ handling defects and dysfunctional contractility, altered global transcriptomic and proteomic profiles, and resulted in apoptosis and cell death. The antioxidant N-acetyl-L-cysteine attenuated these genomic, cellular, and functional alterations. In addition, several other signaling pathways including the p53 and transforming growth factor-ß signaling pathways were also implicated in melphalan-induced cardiotoxicity according to the proteomic and transcriptomic analyses. CONCLUSIONS: Melphalan induces cardiotoxicity through the oxidative stress pathway. This study provides a unique resource of the global transcriptomic and proteomic datasets for melphalan-induced cardiotoxicity and can potentially open up new clinical mechanism-based targets to prevent and treat melphalan-induced cardiotoxicity.


Assuntos
Células-Tronco Pluripotentes Induzidas , Cardiotoxicidade/genética , Células Cultivadas , Humanos , Melfalan/metabolismo , Melfalan/toxicidade , Miócitos Cardíacos/metabolismo , Estresse Oxidativo , Proteômica
3.
Mater Sci Eng C Mater Biol Appl ; 111: 110759, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32279799

RESUMO

Ovarian cancer is considered to be the most fatal reproductive cancers. Melphalan is used to treat ovarian cancer as an intraperitoneal chemotherapy agent. However, elucidating its pharmacokinetic behavior and preparing it for administration are challenging since it undergoes spontaneous hydrolysis. In this study, melphalan is transformed into a macromolecular prodrug by copolymerizing with p-dioxanone. The hydrophobicity of copolymer chains protects melphalan from hydrolysis. Poly(p-dioxanone-co-melphalan; PDCM) is electrosprayed and converted into nanoparticles (PDCM NPs) with diameters of ~300-350 nm to facilitate its intracellular delivery. UPLC-MS and HPLC are applied to verify and monitor the release of melphalan from PDCM NPs. PDCM NPs could suppress the proliferation of SKOV-3 cells. The IC50 of 4.3% melphalan-containing PDCM-3 NP was 70 mg/L, 72 h post administration. These suppression characteristics not only affected by the degradation and then the extracellular release of melphalan from PDCM NPs, but also the uptake via phagocytosis phenomenon in SKOV-3 cells. As revealed by flow cytometry, phagocytosis is a first-order process. Once phagocytosed, PDCM NPs are digested by lysosomes, causing a rapid release of melphalan into the cytoplasm, which ultimately causes suppression of SKOV-3 cell proliferation. Finally, the in vivo antitumor effects of PDCM NPs are verified in xenograft ovarian carcinoma. After a 20-day treatment, the tumor growth rate of the PDCM-3 NP group was (266 ± 178%) which was lower than those in the free melphalan group (367 ± 150%) and control group (648 ± 149%). Besides, significant tissue necrosis and growth suppression were observed in animals administered injections of PDCM NPs. Furthermore, the in vivo tracing results of Nile red-labeled PDCM NPs demonstrated that PDCM-3 NPs might be phagocytosed by macrophages and then taken to adjacent lymph nodes, which is a way of prevention or early treatment of lymphatic metastasis of tumors.


Assuntos
Dioxanos/química , Melfalan/química , Nanopartículas/química , Polímeros/química , Pró-Fármacos/química , Animais , Linhagem Celular Tumoral , Portadores de Fármacos/química , Feminino , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Melfalan/metabolismo , Melfalan/uso terapêutico , Camundongos , Camundongos Nus , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Tamanho da Partícula , Fagocitose , Pró-Fármacos/metabolismo , Pró-Fármacos/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
4.
J Chem Inf Model ; 59(12): 5183-5197, 2019 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-31725294

RESUMO

Adenosine A3 receptor (A3R) is a promising drug target cancer and for a number of other conditions like inflammatory diseases, including asthma and rheumatoid arthritis, glaucoma, chronic obstructive pulmonary disease, and ischemic injury. Currently, there is no experimentally determined structure of A3R. We explored the binding profile of O4-{[3-(2,6-dichlorophenyl)-5-methylisoxazol-4-yl]carbonyl}-2-methyl-1,3-thiazole-4-carbohydroximamide (K18), which is a new specific and competitive antagonist at the orthosteric binding site of A3R. MD simulations and MM-GBSA calculations of the WT A3R in complex with K18 combined with in vitro mutagenic studies show that the most plausible binding conformation for the dichlorophenyl group of K18 is oriented toward trans-membrane helices (TM) 5, 6 and reveal important residues for binding. Further, MM-GBSA calculations distinguish mutations that reduce or maintain or increase antagonistic activity. Our studies show that selectivity of K18 toward A3R is defined not only by direct interactions with residues within the orthosteric binding area but also by remote residues playing a significant role. Although V1695.30 is considered to be a selectivity filter for A3R binders, when it was mutated to glutamic acid, K18 maintained antagonistic potency, in agreement with our previous results obtained for agonists binding profile investigation. Mutation of the direct interacting residue L903.32 in the low region and the remote L2647.35 in the middle/upper region to alanine increases antagonistic potency, suggesting an empty space in the orthosteric area available for increasing antagonist potency. These results approve the computational model for the description of K18 binding at A3R, which we previously performed for agonists binding to A3R, and the design of more effective antagonists based on K18.


Assuntos
Antagonistas do Receptor A3 de Adenosina/farmacologia , Simulação de Dinâmica Molecular , Mutagênese , Receptor A3 de Adenosina/metabolismo , Antagonistas do Receptor A3 de Adenosina/química , Antagonistas do Receptor A3 de Adenosina/metabolismo , Amidas/química , Amidas/metabolismo , Amidas/farmacologia , Melfalan/metabolismo , Melfalan/farmacologia , Simulação de Acoplamento Molecular , Distribuição de Poisson , Ligação Proteica , Conformação Proteica , Receptor A3 de Adenosina/química , Receptor A3 de Adenosina/genética , Especificidade por Substrato , Termodinâmica , gama-Globulinas/metabolismo , gama-Globulinas/farmacologia
5.
Bioorg Chem ; 84: 418-433, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30554081

RESUMO

1,5-Dideoxy-1,5-imino-l-fucitol (1-deoxyfuconojirimycin, DFJ) is an iminosugar that inhibits fucosidases. Herein, N-alkyl DFJs have been synthesised and tested against the α-fucosidases of T. maritima (bacterial origin) and B. taurus (bovine origin). The N-alkyl derivatives were inactive against the bacterial fucosidase, while inhibiting the bovine enzyme. Docking of inhibitors to homology models, generated for the bovine and human fucosidases, was carried out. N-Decyl-DFJ was toxic to cancer cell lines and was more potent than the other N-alkyl DFJs studied.


Assuntos
Inibidores Enzimáticos/síntese química , Álcoois Açúcares/química , alfa-L-Fucosidase/antagonistas & inibidores , 1-Desoxinojirimicina/análogos & derivados , 1-Desoxinojirimicina/química , 1-Desoxinojirimicina/metabolismo , Bactérias/enzimologia , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , Imunoglobulina G/metabolismo , Imunoglobulina G/farmacologia , Concentração Inibidora 50 , Melfalan/síntese química , Melfalan/metabolismo , Melfalan/farmacologia , Simulação de Acoplamento Molecular , Relação Estrutura-Atividade , Álcoois Açúcares/metabolismo , alfa-L-Fucosidase/metabolismo
6.
Mol Pharm ; 14(11): 3772-3779, 2017 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-28969419

RESUMO

Tumor necrosis factor-α has shown potent antitumor effects in preclinical and clinical studies. However, severe side effects at less than therapeutic doses have limited its systemic delivery, prompting the need for a new strategy for targeted delivery of the protein to tumors. Here, we report a fusion protein of mouse tumor necrosis factor (TNF)-α (mTNFα) and a cancer-targeting, high-affinity aptide and investigate its therapeutic efficacy in tumor-bearing mice. A fusion protein consisting of mTNFα, a linker, and an aptide specific to extra domain B (EDB) of fibronectin (APTEDB), designated mTNFα-APTEDB, was successfully produced by expression in Escherichia coli. mTNFα-APTEDB retained specificity and affinity for its target, EDB. In mice bearing EDB-overexpressing fibrosarcomas, mTNFα-APTEDB showed greater efficacy in inhibiting tumor growth than mTNFα alone or mTNFα linked to a nonrelevant aptide, without causing an appreciable loss in body weight. Moreover, in vivo antitumor efficacy was further significantly increased by combination treatment with the chemotherapeutic drug, melphalan, suggesting a synergistic effect attributable to enhanced drug uptake into the tumor as a result of TNFα-mediated enhanced vascular permeability. These results suggest that a fusion protein of mTNFα with a cancer-targeting peptide could be a new anticancer therapeutic option for ensuring potent antitumor efficacy after systemic delivery.


Assuntos
Fibronectinas/metabolismo , Peptídeos/metabolismo , Proteínas Recombinantes de Fusão/uso terapêutico , Fator de Necrose Tumoral alfa/metabolismo , Animais , Fibronectinas/química , Fibrossarcoma/tratamento farmacológico , Melfalan/química , Melfalan/metabolismo , Camundongos , Peptídeos/química , Proteínas Recombinantes de Fusão/metabolismo , Fator de Necrose Tumoral alfa/química
7.
Pharm Res ; 33(8): 1913-22, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27091031

RESUMO

PURPOSE: Busulfan-melphalan high-dose chemotherapy followed by autologous stem cell transplantation is an essential consolidation treatment of high-risk neuroblastoma in children. Main treatment limitation is hepatic veno-occlusive disease, the most severe and frequent extra-hematological toxicity. This life threatening toxicity has been related to a drug interaction between busulfan and melphalan which might be increased by prior disturbance of iron homeostasis, i.e. an increased plasma ferritin level. METHODS: We performed an experimental study of busulfan and melphalan pharmacodynamic and pharmacokinetics in iron overloaded mice. RESULTS: Iron excess dramatically increased the toxicity of melphalan or busulfan melphalan combination in mice but it did not modify the clearance of either busulfan or melphalan. We show that prior busulfan treatment impairs the clearance of melphalan. This clearance alteration was exacerbated in iron overloaded mice demonstrating a pharmacokinetic interaction. Additionally, iron overload increased melphalan toxicity without altering its pharmacokinetics, suggesting a pharmacodynamic interaction between iron and melphalan. Based on iron homeostasis disturbance, we postulated that prior induction of ferritin, through Nrf2 activation after oxidative stress, may be associated with the alteration of melphalan metabolism. CONCLUSION: Iron overload increases melphalan and busulfan-melphalan toxicity through a pharmacodynamic interaction and reveals a pharmacokinetic drug interaction between busulfan and melphalan.


Assuntos
Bussulfano/metabolismo , Bussulfano/toxicidade , Sobrecarga de Ferro/metabolismo , Melfalan/metabolismo , Melfalan/toxicidade , Animais , Antineoplásicos Alquilantes/metabolismo , Antineoplásicos Alquilantes/toxicidade , Interações Medicamentosas/fisiologia , Sobrecarga de Ferro/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
8.
J Neurosci ; 35(40): 13761-72, 2015 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-26446227

RESUMO

The olfactory epithelium (OE) is one of the few tissues to undergo constitutive neurogenesis throughout the mammalian lifespan. It is composed of multiple cell types including olfactory sensory neurons (OSNs) that are readily replaced by two populations of basal stem cells, frequently dividing globose basal cells and quiescent horizontal basal cells (HBCs). However, the precise mechanisms by which these cells mediate OE regeneration are unclear. Here, we show for the first time that the HBC subpopulation of basal stem cells uniquely possesses primary cilia that are aligned in an apical orientation in direct apposition to sustentacular cell end feet. The positioning of these cilia suggests that they function in the detection of growth signals and/or differentiation cues. To test this idea, we generated an inducible, cell type-specific Ift88 knock-out mouse line (K5rtTA;tetOCre;Ift88(fl/fl)) to disrupt cilia formation and maintenance specifically in HBCs. Surprisingly, the loss of HBC cilia did not affect the maintenance of the adult OE but dramatically impaired the regeneration of OSNs following lesion. Furthermore, the loss of cilia during development resulted in a region-specific decrease in neurogenesis, implicating HBCs in the establishment of the OE. Together, these results suggest a novel role for primary cilia in HBC activation, proliferation, and differentiation. SIGNIFICANCE STATEMENT: We show for the first time the presence of primary cilia on a quiescent population of basal stem cells, the horizontal basal cells (HBCs), in the olfactory epithelium (OE). Importantly, our data demonstrate that cilia on HBCs are necessary for regeneration of the OE following injury. Moreover, the disruption of HBC cilia alters neurogenesis during the development of the OE, providing evidence that HBCs participate in the establishment of this tissue. These data suggest that the mechanisms of penetrance for ciliopathies in the OE extend beyond that of defects in olfactory sensory neurons and may include alterations in OE maintenance and regeneration.


Assuntos
Cílios/genética , Mucosa Olfatória/lesões , Regeneração/genética , Fatores de Ribosilação do ADP/genética , Animais , Animais Recém-Nascidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Doxiciclina/administração & dosagem , Embrião de Mamíferos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Histona Desmetilases/metabolismo , Melfalan/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Proteína de Marcador Olfatório/metabolismo , Mucosa Olfatória/citologia , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo , gama-Globulinas/metabolismo
9.
J Neurooncol ; 122(3): 517-27, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25800004

RESUMO

The tyrosine kinase receptor c-Met has been suggested to be involved in crucial parts of glioma biology like tumor stemness, growth and invasion. The aim of this study was to investigate the prognostic value of c-Met in a population-based glioma patient cohort. Tissue samples from 238 patients with WHO grade I, II, III and IV tumors were analyzed using immunohistochemical staining and advanced image analysis. Strong c-Met expression was found in tumor cells, blood vessels, and peri-necrotic areas. At the subcellular level, c-Met was identified in the cytoplasm and in the cell membrane. Measurements of high c-Met intensity correlated with high WHO grade (p = 0.006) but no association with survival was observed in patients with WHO grade II (p = 0.09) or III (p = 0.17) tumors. High expression of c-Met was associated with shorter overall survival in patients with glioblastoma multiforme (p = 0.03). However the prognostic effect of c-Met in glioblastomas was time-dependent and only observed in patients who survived more than 8.5 months, and not within the first 8.5 months after diagnosis. This was significant in multivariate analysis (HR 1.99, 95 % CI 1.29-3.08, p = 0.002) adjusted for treatment and the clinical variables age (HR 1.01, 95 % CI 0.99-1.03, p = 0.30), performance status (HR 1.34, 95 % CI 1.17-1.53, p < 0.001), and tumor crossing midline (HR 1.28, 95 % CI 0.79-2.07, p = 0.29). In conclusion, this study showed that high levels of c-Met holds unfavorable prognostic value in glioblastomas.


Assuntos
Neoplasias Encefálicas/metabolismo , Cefalosporinas/metabolismo , Glioblastoma/metabolismo , Melfalan/análogos & derivados , Neoplasias Encefálicas/diagnóstico , Linhagem Celular Tumoral , Estudos de Coortes , Planejamento em Saúde Comunitária , Feminino , Glioblastoma/diagnóstico , Humanos , Estimativa de Kaplan-Meier , Imageamento por Ressonância Magnética , Masculino , Melfalan/metabolismo , Pessoa de Meia-Idade , Modelos de Riscos Proporcionais , Índice de Gravidade de Doença
10.
Biochim Biophys Acta ; 1848(5): 1099-110, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25646577

RESUMO

Recently, we showed that tetrasaccharide selectin ligand SiaLeX provided targeted delivery of liposomes loaded in the bilayer with melphalan lipophilic prodrug to tumour endothelium followed by severe injury of tumour vessels in a Lewis lung carcinoma model. Here, we study the impact of SiaLeX ligand on the interactions of liposomes with human umbilical vein endothelial cells (HUVEC) using flow cytometry, spectrofluorimetry and confocal microscopy. Liposomes composed of egg phosphatidylcholine/yeast phosphatidylinositol/1,2-dioleoyl glycerol ester of melphalan, 8:1:1, by mol, and varying percentages of lipophilic SiaLeX conjugate were labelled with BODIPY-phosphatidylcholine. The increase in SiaLeX content in liposomes led to a proportional increase in their uptake by cytokine-activated cells as opposed to non-activated HUVEC: for 10% SiaLeX liposomes, binding avidity and overall accumulation increased 14- and 6-fold, respectively. The early stages of intracellular traffic of targeted liposomes in the activated cells were monitored by co-localisation with the trackers of organelles. Endocytosis of SiaLeX liposomes occurred mostly via clathrin-independent pathways, which does not contradict the available literature data on E-selectin localisation in the plasma membrane. Using dual fluorescence labelling, with rhodamine-labelled phospholipid and calcein encapsulated at self-quenching concentrations, we found that SiaLeX liposomes undergo rapid (within minutes) internalisation by activated HUVEC accompanied by the disruption of liposomes; non-activated cells consumed a negligible dose of liposomes during at least 1.5h. Our data evidence the selective effect of SiaLeX formulations on activated endothelial cells and indicate their potential for intracellular delivery of melphalan lipophilic prodrug.


Assuntos
Antineoplásicos Alquilantes/metabolismo , Portadores de Fármacos , Endocitose , Células Endoteliais da Veia Umbilical Humana/metabolismo , Antígenos CD15/metabolismo , Lipídeos/química , Melfalan/metabolismo , Antineoplásicos Alquilantes/química , Células Cultivadas , Química Farmacêutica , Diglicerídeos/química , Relação Dose-Resposta a Droga , Selectina E/metabolismo , Endocitose/efeitos dos fármacos , Citometria de Fluxo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Cinética , Antígenos CD15/química , Lipossomos , Melfalan/análogos & derivados , Melfalan/química , Microscopia Confocal , Fosfatidilcolinas/química , Fosfatidilinositóis/química , Antígeno Sialil Lewis X , Espectrometria de Fluorescência , Fator de Necrose Tumoral alfa/farmacologia
11.
Am J Physiol Renal Physiol ; 308(4): F330-8, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25477469

RESUMO

Chronic lymphatic leukemia (CLL) is often associated with nephritic syndrome. Effective treatment of CLL by chlorambucil and bendamustine leads to the restoration of renal function. In this contribution, we sought to elucidate the impact of organic anion transporters (OATs) on the uptake of bendamustine and chlorambucil as a probable reason for the superior efficacy of bendamustine over chlorambucil in the treatment of CLL. We examined the effects of structural analogs of p-aminohippurate (PAH), melphalan, chlorambucil, and bendamustine, on OAT1-mediated [(3)H]PAH uptake and OAT3- and OAT4-mediated [(3)H]estrone sulfate (ES) uptake in stably transfected human embryonic kidney-293 cells. Melphalan had no significant inhibitory effect on any OAT, whereas chlorambucil reduced OAT1-, OAT3-, and OAT4-mediated uptake of PAH or ES down to 14.6%, 16.3%, and 66.0% of control, respectively. Bendamustine inhibited only OAT3-mediated ES uptake, which was reduced down to 14.3% of control cells, suggesting that it interacts exclusively with OAT3. The IC50 value for OAT3 was calculated to be 0.8 µM. Real-time PCR experiments demonstrated a high expression of OAT3 in lymphoma cell lines as well as primary CLL cells. OAT3-mediated accumulation of bendamustine was associated with reduced cell proliferation and an increased rate of apoptosis. We conclude that the high efficacy of bendamustine in treating CLL might be partly contributed to the expression of OAT3 in lymphoma cells and the high affinity of bendamustine for this transporter.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Leucemia Linfocítica Crônica de Células B/metabolismo , Linfoma de Células T/metabolismo , Compostos de Mostarda Nitrogenada/farmacologia , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Antineoplásicos Alquilantes/metabolismo , Apoptose/efeitos dos fármacos , Cloridrato de Bendamustina , Proliferação de Células/efeitos dos fármacos , Clorambucila/metabolismo , Clorambucila/farmacologia , Relação Dose-Resposta a Droga , Estrona/análogos & derivados , Estrona/metabolismo , Células HEK293 , Humanos , Células Jurkat , Leucemia Linfocítica Crônica de Células B/genética , Leucemia Linfocítica Crônica de Células B/patologia , Linfoma de Células T/genética , Linfoma de Células T/patologia , Melfalan/metabolismo , Melfalan/farmacologia , Compostos de Mostarda Nitrogenada/metabolismo , Proteína 1 Transportadora de Ânions Orgânicos/antagonistas & inibidores , Proteína 1 Transportadora de Ânions Orgânicos/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/antagonistas & inibidores , Transportadores de Ânions Orgânicos Sódio-Independentes/genética , Transfecção , Células Tumorais Cultivadas , Ácido p-Aminoipúrico/metabolismo
12.
Vopr Onkol ; 61(6): 932-6, 2015.
Artigo em Russo | MEDLINE | ID: mdl-26995981

RESUMO

The study of pharmacokinetics of melphalan in the perfusate and blood plasma during isolated limb regional perfusion (ILRP) was carried out in patients with melanoma (n=21) and soft tissue sarcoma (n = 24). Melphalan was administered as 10 mg/l for a lower extremity and 13 mg/l for a upper extremity. Quantification of melphalan in perfusate and blood samples was performed by means of liquid chromatography/tandem mass spectrometry. 30 samples of the perfusate and 27 venous blood samples were analyzed. During the first 5 minutes of ILRP concentration of melphalan in the perfusate decreased to 13.2% of the initial value, and by the end of perfusion (60 minutes) it was 3.3%. The amount of melphalan in the blood plasma of the patients by the end of ILRP wasn't higher than 1.6% from the administered dose. That demonstrates minor systemic absorption of the drug during ILRP. Moreover melphalan concentration in the blood plasma during the perfusion was in average 0.015-0.223 mg/l which is significantly lower compared to the blood plasma concentrations after intravenous administration of melphalan. Thus ILRP procedure provided 97% of the melphalan dose accumulation in the soft tissues of a limb and in tumor tissues. Also pharmacokinetic advantage of melphalan over systemic administration of the drug was shown.


Assuntos
Antineoplásicos Alquilantes/farmacocinética , Quimioterapia do Câncer por Perfusão Regional , Melanoma/tratamento farmacológico , Melfalan/farmacocinética , Sarcoma/tratamento farmacológico , Neoplasias Cutâneas/tratamento farmacológico , Adulto , Idoso , Antineoplásicos Alquilantes/administração & dosagem , Antineoplásicos Alquilantes/sangue , Antineoplásicos Alquilantes/metabolismo , Cromatografia Líquida , Esquema de Medicação , Feminino , Humanos , Infusões Intravenosas , Masculino , Melanoma/sangue , Melanoma/metabolismo , Melfalan/administração & dosagem , Melfalan/sangue , Melfalan/metabolismo , Pessoa de Meia-Idade , Sarcoma/sangue , Sarcoma/metabolismo , Neoplasias Cutâneas/sangue , Neoplasias Cutâneas/metabolismo , Espectrometria de Massas em Tandem , Melanoma Maligno Cutâneo
13.
CNS Neurosci Ther ; 20(10): 916-29, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24992648

RESUMO

AIMS: Different trophic factors are known to promote retinal ganglion cell survival and regeneration, but each had their own limitations. We report that hepatocyte growth factor (HGF) confers distinct advantages in supporting ganglion cell survival and axonal regeneration, when compared to two well-established trophic factors ciliary neurotrophic factor (CNTF) and brain-derived neurotrophic factor (BDNF). METHODS: Ganglion cells in adult hamster were injured by cutting the optic nerve. HGF, CNTF, or BDNF was injected at different dosages intravitreally after injury. Ganglion cell survival was quantified at 7, 14, or 28 days postinjury. Peripheral nerve (PN) grafting to the cut optic nerve of the growth factor-injected eye was performed either immediately after injury or delayed until 7 days post-injury. Expression of heat-shock protein 27 and changes in microglia numbers were quantified in different growth factor groups. The cellular distribution of c-Met in the retina was examined by anti-c-Met immunostaining. RESULTS: Hepatocyte Growth Factor (HGF) was equally potent as BDNF in promoting short-term survival (up to 14 days post-injury) and also supported survival at 28 days post-injury when ganglion cells treated by CNTF or BDNF failed to be sustained. When grafting was performed without delay, HGF stimulated twice the number of axons to regenerate compared with control but was less potent than CNTF. However, in PN grafting delayed for 7 days after optic nerve injury, HGF maintained a better propensity of ganglion cells to regenerate than CNTF. Unlike CNTF, HGF application did not increase HSP27 expression in ganglion cells. Microglia proliferation was prolonged in HGF-treated retinas compared with CNTF or BDNF. C-Met was localized to both ganglion cells and Muller cells, suggesting HGF could be neuroprotective via interacting with both neurons and glia. CONCLUSION: Compared with CNTF or BDNF, HGF is advantageous in sustaining long-term ganglion cell survival and their propensity to respond to favorable stimuli.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Fator Neurotrófico Ciliar , Fator de Crescimento de Hepatócito , Regeneração Nervosa/efeitos dos fármacos , Traumatismos do Nervo Óptico , Células Ganglionares da Retina/efeitos dos fármacos , Animais , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Fator Neurotrófico Derivado do Encéfalo/uso terapêutico , Sobrevivência Celular , Cefalosporinas/metabolismo , Fator Neurotrófico Ciliar/farmacologia , Fator Neurotrófico Ciliar/uso terapêutico , Cricetinae , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Proteínas de Choque Térmico HSP27/metabolismo , Fator de Crescimento de Hepatócito/farmacologia , Fator de Crescimento de Hepatócito/uso terapêutico , Melfalan/análogos & derivados , Melfalan/metabolismo , Mesocricetus , Traumatismos do Nervo Óptico/tratamento farmacológico , Traumatismos do Nervo Óptico/patologia , Traumatismos do Nervo Óptico/fisiopatologia , Fatores de Tempo
14.
Carbohydr Polym ; 98(1): 36-42, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-23987314

RESUMO

A series of melphalan-O-carboxymethyl chitosan (Mel-OCM-chitosan) conjugates with different spacers were prepared and structurally characterized. All conjugates showed satisfactory water-solubility (160-217 times of Mel solubility). In vitro drug release behaviors by both chemical and enzymatic hydrolysis were investigated. The prodrugs released Mel rapidly within papain and lysosomal enzymes of about 40-75%, while released only about 4-5% in buffer and plasma, which suggested that the conjugates have good plasma stability and the hydrolysis in both papain and lysosomes occurs mostly via enzymolysis. It was found that the spacers have important effect on the drug content, water solubility, drug release properties and cytotoxicity of Mel-OCM-chitosan conjugates. Cytotoxicity studies by MTT assay demonstrated that these conjugates had 52-70% of cytotoxicity against RPMI8226 cells in vitro as compared with free Mel, indicating the conjugates did not lose anti-cancer activity of Mel. Overall these studies indicated Mel-OCM-chitosan conjugates as potential prodrugs for cancer treatment.


Assuntos
Antineoplásicos/química , Quitosana/análogos & derivados , Portadores de Fármacos/química , Melfalan/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Quitosana/química , Estabilidade de Medicamentos , Humanos , Melfalan/metabolismo , Melfalan/farmacocinética , Melfalan/farmacologia , Pró-Fármacos/metabolismo , Solubilidade , Água/química
15.
Cancer Chemother Pharmacol ; 69(2): 397-405, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21789689

RESUMO

PURPOSE: To evaluate the ability of a physiology-based pharmacokinetic (PBPK) model to predict the systemic drug exposure of high- and low-dose etoposide in children from a model developed with adult data. METHODS: Simulations were performed with PK-Sim(®) (Bayer Technology Services). Model development was done using data from adult patients receiving etoposide in a conventional and high-dose polychemotherapy regimen before stem cell transplantation. Michaelis-Menten parameters from in vitro experiments reported in the literature were applied to describe the metabolism and excretion processes by P450 enzymes and transporters. The model was scaled down to children and compared to etoposide plasma concentrations in this age group. RESULTS: Simulated plasma concentration-time courses of protein-bound and free etoposide in adults for high- and low-dose schedules agreed with the observed data. Mean simulated total clearance of high- and low-dose etoposide was 0.70 ml/min/kg (Cl(observed): 0.70 ml/min/kg) versus 0.50 ml/min/kg (Cl(observed): 0.60 ml/min/kg), respectively. Integrated Michaelis-Menten kinetics was adequately transformed to age-related pharmacokinetics in children. Predictions of the pharmacokinetics in different age groups were also in good agreement with observed data. Drug interactions triggered by P-glycoprotein inhibitors or nephrotoxic drugs can also be elucidated. CONCLUSIONS: The PBPK model matched the pharmacokinetics in different dosing regimens in adults. Furthermore, the scaling procedure from the adult model to children provides useful predictions for paediatric patients. Comedication with drugs influencing the metabolism and excretion has to be taken into account. This approach could be useful for planning pharmacokinetic studies in children.


Assuntos
Antineoplásicos Fitogênicos/farmacocinética , Etoposídeo/farmacocinética , Modelos Biológicos , Neoplasias/metabolismo , Adolescente , Adulto , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Bussulfano/administração & dosagem , Bussulfano/metabolismo , Bussulfano/farmacocinética , Carboplatina/administração & dosagem , Carboplatina/metabolismo , Carboplatina/farmacocinética , Criança , Pré-Escolar , Simulação por Computador , Ciclofosfamida/administração & dosagem , Ciclofosfamida/metabolismo , Ciclofosfamida/farmacocinética , Citocromo P-450 CYP3A/metabolismo , Relação Dose-Resposta a Droga , Esquema de Medicação , Interações Medicamentosas , Etoposídeo/administração & dosagem , Etoposídeo/metabolismo , Feminino , Humanos , Lactente , Masculino , Melfalan/administração & dosagem , Melfalan/metabolismo , Melfalan/farmacocinética , Taxa de Depuração Metabólica , Pessoa de Meia-Idade , Neoplasias/tratamento farmacológico , Neoplasias/fisiopatologia , Ligação Proteica
16.
Bioconjug Chem ; 21(10): 1728-43, 2010 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-20849062

RESUMO

Bendamustine and melphalan are very promising alkylating drugs with applicability in the treatment of various tumoral diseases, e.g., chronic lymphocytic leukemia (CLL) or breast cancer. However, numerous adverse effects limited their use. Therefore, 1,3,5-tris(3-aminopropyl)benzene (G0) and its G1 analogue 3,5-bis(3-aminopropyl)-N-(3-{3,5-bis[3-{3,5-bis(3-aminopropyl)benzoylamino}propyl]phenyl}propyl)benzamide were selected to design cytostatic drug-dendrimer conjugates to achieve tumor cell accumulation by endocytosis as already demonstrated for platinum complexes. The dendrimers act as carriers and an N-(2-hydroxyethyl)maleimide spacer between drug and carrier should guarantee a selective release of the cytostatics in the tumor cells. The resulting cytotoxicity was determined in vitro using the human MCF-7 and MDA-MB-231 breast cancer cell lines. It was demonstrated that melphalan caused cytotoxic effects depending on its free amino group (Boc protection strongly decreased the activity) but independent of a derivation of the carboxylic group (dendrimers and spacer binding). Esterification of bendamustine with the N-(2-hydroxyethyl)maleimide spacer strongly increased the hydrolytic stability of the N-lost moiety, so antiproliferative effects were yet observed in vitro.


Assuntos
Antineoplásicos Alquilantes/síntese química , Antineoplásicos Alquilantes/farmacologia , Dendrímeros/química , Melfalan/química , Melfalan/farmacologia , Nanoconjugados/química , Compostos de Mostarda Nitrogenada/química , Compostos de Mostarda Nitrogenada/farmacologia , Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/metabolismo , Cloridrato de Bendamustina , Linhagem Celular Tumoral , Dendrímeros/síntese química , Endocitose , Humanos , Concentração Inibidora 50 , Melfalan/metabolismo , Modelos Moleculares , Conformação Molecular , Compostos de Mostarda Nitrogenada/metabolismo , Tamanho da Partícula , Permeabilidade
17.
J Chromatogr B Analyt Technol Biomed Life Sci ; 877(27): 3089-96, 2009 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-19674945

RESUMO

Two high-performance liquid chromatography tandem mass spectrometry (LC-MS/MS) assays are described for the quantification of melphalan in human plasma. N-phenyldiethanolamine was tested as internal standard. The first assay consisted of a protein precipitation by cold methanol and a reversed-phase HPLC whereas the second one was based on a solid phase extraction and a hydrophilic interaction chromatography. Both provided a very satisfactory mean extraction yield with a small volume of sample. The first method was simple, rapid and used as a routine assay. The second one was developed in order to determine melphalan hydrolysis products and to avoid scarce cases when interferences from biological matrix alter the quantification of melphalan using the first method. The two assays were linear and sensitive in the range of 1-500ng/mL for the first one and in a range of 25-2000ng/mL for the second one. Concentrations out of the range fixed with the first method were also validated. The procedure was reliable with precision and accuracy below 10%. All compounds were detected after positive mode electrospray ionization in selected reaction monitoring mode. These new analytical procedures were developed for melphalan pharmacokinetic studies or therapeutic drug monitoring.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Melfalan/sangue , Espectrometria de Massas em Tandem/métodos , Idoso , Estabilidade de Medicamentos , Humanos , Hidrólise , Análise dos Mínimos Quadrados , Masculino , Melfalan/metabolismo , Melfalan/farmacocinética , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Extração em Fase Sólida/métodos
18.
Anal Bioanal Chem ; 392(5): 805-17, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18622599

RESUMO

Melphalan is a bifunctional alkylating agent that covalently binds to the nucleophilic sites present in DNA. In this study we investigated oligonucleotides prepared enzymatically from DNA modified with melphalan. Calf thymus DNA was incubated in-vitro with melphalan and the resulting modifications were enzymatically cleaved by means of benzonase and nuclease S1. Efficient sample preconcentration was achieved by solid-phase extraction, in which phenyl phase cartridges resulted in better recovery of the modified species than C(18). The applied enzymatic digestion time resulted in production of trinucleotide adducts which were efficiently separated and detected by use of reversed-phase HPLC coupled to an ion-trap mass spectrometer with electrospray ionization. It was assumed that melphalan could act as both a monofunctional and bifunctional alkylating agent. Mono-alkylated adducts were much more abundant, however, and the alkylation site was located on the nucleobases. On the other hand, we unequivocally identified cross-link formation in DNA, even though at low abundance and only a few adduct types were detected.


Assuntos
Antineoplásicos Alquilantes/isolamento & purificação , Cromatografia Líquida de Alta Pressão/métodos , Adutos de DNA/isolamento & purificação , DNA/metabolismo , Desoxirribonucleotídeos/isolamento & purificação , Melfalan/isolamento & purificação , Espectrometria de Massas por Ionização por Electrospray/métodos , Alquilação , Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/metabolismo , Aziridinas/química , Reagentes de Ligações Cruzadas/química , Adutos de DNA/química , Adutos de DNA/metabolismo , Desoxirribonucleotídeos/química , Desoxirribonucleotídeos/metabolismo , Endodesoxirribonucleases/metabolismo , Endorribonucleases/metabolismo , Melfalan/química , Melfalan/metabolismo , Compostos de Mostarda Nitrogenada/metabolismo , Endonucleases Específicas para DNA e RNA de Cadeia Simples/metabolismo , Fatores de Tempo
19.
Cytometry A ; 71(11): 951-60, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17879237

RESUMO

The F7-26 monoclonal antibody (Mab) has been reported to be specific for single-strand DNA damage (ssDNA) and to also identify cells in apoptosis. We carriedout studies to determine if F7-26 binding measured by flow cytometry was able to specifically identify exogenous ssDNA as opposed to DNA damage from apoptosis. Neuroblastoma cells were treated with melphalan (L-PAM), fenretinide, 4-hydroperoxycyclophosphamide (4-HC)+/-pan-caspase inhibitor BOC-d-fmk, topotecan or with 10Gy gamma radiation+/-hydrogen peroxide (H2O2) and fixed immediately postradiation. Cytotoxicity was measured by DIMSCAN digital imaging fluorescence assay. The degree of ssDNA damage was analyzed by flow cytometry using Mab F7-26, with DNA visualized by propidium iodide counterstaining. Flow cytometry was used to measure apoptosis detected by terminal deoxynucleotidyltransferase (TUNEL) assay and reactive oxygen species (ROS) by carboxy-dichlorofluorescein diacetate. Irradiated and immediately fixed neuroblastoma cells showed increased ssDNA, but not apoptosis by TUNEL (TUNEL-negative). 4-HC or L-PAM+/-BOC-d-fmk increased ssDNA (F7-26-positive), but BOC-d-fmk prevented TUNEL staining. Fenretinide increased apoptosis by TUNEL but not ssDNA damage detected with F7-26. Enhanced ssDNA in neuroblastoma cells treated with radiation+H2O2 was associated with increased ROS. Topotecan increased both ssDNA and cytotoxicity in 4-HC-treated cells. These data demonstrate that Mab F7-26 recognized ssDNA due to exogenous DNA damage, rather than apoptosis. This assay should be useful to characterize the mechanism of action of antineoplastic drugs.


Assuntos
Anticorpos Monoclonais/metabolismo , Dano ao DNA , DNA de Cadeia Simples , Citometria de Fluxo/métodos , Antineoplásicos/metabolismo , Apoptose/fisiologia , Compostos de Benzil/metabolismo , Linhagem Celular Tumoral/metabolismo , Linhagem Celular Tumoral/efeitos da radiação , Criança , DNA de Cadeia Simples/química , DNA de Cadeia Simples/metabolismo , Inibidores Enzimáticos/metabolismo , Fenretinida/metabolismo , Raios gama , Humanos , Hidrocarbonetos Fluorados/metabolismo , Peróxido de Hidrogênio/metabolismo , Marcação In Situ das Extremidades Cortadas , Melfalan/metabolismo , Neuroblastoma/genética , Oxidantes/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Topotecan/metabolismo
20.
Eur J Pharm Biopharm ; 67(3): 752-8, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17560100

RESUMO

The therapeutic efficacy of prophalan-L, the L-proline prodrug of melphalan that demonstrated prolidase-dependent bioactivation to melphalan, was examined in vivo in a mouse melanoma model. Prophalan-L exhibited 2- to 2.5-fold higher hydrolytic and cytotoxic activity than prophalan-D, the D-analog, in B16-F10 murine melanoma cells in vitro. Prophalan-L cytotoxicity in B16-F10 cells was lower (GI50=221 microM) than that of melphalan (GI50=173 microM). The tumor growth profiles in C57BL/6J mice injected with B16-F10 cells and treated with melphalan (5.5 microg/g i.p.) and equimolar concentrations of the prodrugs demonstrated significant difference between the control (buffered saline) and melphalan or prophalan-L but no significant difference between control and prophalan-D or between melphalan and prophalan-L. Prophalan-L was significantly less toxic than melphalan, while no significant difference was observed in toxicity, measured as percent weight loss, between the prodrugs and saline control. Tumor reduction efficacy at high doses (12 microg/g i.p.) was similar for melphalan and prophalan-L; however, fatal toxicity was associated with melphalan while prophalan-L exhibited significantly lower systemic toxicity. An excellent correlation between GI50 and tumor reduction efficacy was observed for the tested drugs (r2=0.95). Prophalan-L thus demonstrates higher therapeutic index than melphalan in the murine melanoma model.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Melanoma Experimental/tratamento farmacológico , Melfalan/análogos & derivados , Melfalan/uso terapêutico , Pró-Fármacos/uso terapêutico , Prolina/análogos & derivados , Animais , Antineoplásicos Alquilantes/metabolismo , Peso Corporal/efeitos dos fármacos , Calorimetria , Proliferação de Células/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Feminino , Hidrólise , Indicadores e Reagentes , Melanoma Experimental/patologia , Melfalan/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Pró-Fármacos/metabolismo , Prolina/metabolismo , Prolina/uso terapêutico , Espectrofotometria Ultravioleta , Estereoisomerismo , Análise de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA