Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Org Biomol Chem ; 17(6): 1480-1486, 2019 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-30681115

RESUMO

Prostate cancer represents a major public health threat as it is one of the most common male cancers worldwide. The prostate-specific membrane antigen (PSMA) is highly over-expressed in prostatic cancer cells in a manner that correlates with both tumour stage and clinical outcome. As such, PSMA has been identified as an attractive target for both imaging and treatment of prostate cancer. In recent years the focus on urea-based peptidomimetic inhibitors of the PSMA (representing low molecular weight/high affinity binders) has intensified as they have found use in the clinical imaging of prostate tumours. Reported herein are the design, synthesis and evaluation of a new fluorinated PSMA targeting small-molecule, FDA-PEG-GUL, which possesses the Glu-NH-CO-NH-Lys pharmacophore conjugated to a 5'-fluorodeoxy-adenosine unit. Inhibition assays were performed with FDA-PEG-GUL which revealed that it inhibits the PSMA in the nanomolar range. Additionally, it has been purposely designed so that it can be produced using the fluorinase enzyme from its chlorinated precursor, allowing for the enzymatic synthesis of radiolabelled [18F]FDA-PEG-GUL via a nucleophilic reaction that takes place in experimentally advantageous conditions (in water at neutral pH and at ambient temperature). Specific binding of [18F]FDA-PEG-GUL to PSMA expressing cancer cells was demonstrated, validating it as a promising PSMA diagnostic tool. This work establishes a successful substrate scope expansion for the fluorinase and demonstrates its first application towards targeting the PSMA.


Assuntos
Antígenos de Superfície/metabolismo , Proteínas de Bactérias/metabolismo , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/metabolismo , Radioisótopos de Flúor , Glutamato Carboxipeptidase II/metabolismo , Lisina/química , Oxirredutases/metabolismo , Radioquímica/métodos , Linhagem Celular Tumoral , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Glutamato Carboxipeptidase II/antagonistas & inibidores , Humanos , Marcação por Isótopo , Ligantes , Streptomyces/enzimologia
2.
Bioconjug Chem ; 27(5): 1332-40, 2016 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-27077642

RESUMO

Cyclic CNGRC (cCNGRC) peptides are very important targeting ligands for Aminopeptidase N (APN or CD13), which is overexpressed on the surface of many cancer cells. In this work we have (1) developed an efficient solid-phase synthesis and (2) tested on purified porcine APN and APN-expressing human cells two different classes of cCNGRC peptides: the first carrying a biotin affinity tag or a fluorescent tag attached to the carboxyl Arg-Cys-COOH terminus and the second with the tags attached to the amino H2N-Cys-Asn terminus. Carboxyl-terminus functionalized cCNGRC peptides 3, 6, and 8 showed good affinity for porcine APN and very good capacity to target and be internalized into APN-expressing cells. In contrast, amino-terminus functionalized cCNGRC peptides 4, 5, and 7 displayed significantly decreased affinity and targeting capacity. These results, which are in agreement with the recently reported X-ray structure of a cCNGRC peptide bound to APN showing important stabilizing interactions between the unprotected cCNGRC amino terminus and the APN active site, indicate that the carboxyl and not the amino-terminus of cCNGRC peptides should be used as a "handle" for the attachment of toxic payloads for therapy or isotopically labeled functions for imaging and nuclear medicine.


Assuntos
Antígenos CD13/metabolismo , Oligopeptídeos/química , Oligopeptídeos/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Humanos , Modelos Moleculares , Conformação Proteica , Suínos
3.
Chemistry ; 22(31): 10998-1004, 2016 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-27374143

RESUMO

We report a last-step fluorinase-catalyzed [(18) F]-fluorination of a cysteine-containing RGD peptide. The peptide was attached through sulfur to a modified and more hydrophilic variant of the recently disclosed Barbas linker which was itself linked to a chloroadenosine moiety via a PEGylated chain. The fluorinase was able to use this construct as a substrate for a transhalogenation reaction to generate [(18) F]-radiolabeled RGD peptides, which retained high affinity to cancer-cell relevant αv ß3 integrins.


Assuntos
Cisteína/química , Radioisótopos de Flúor/química , Humanos , Modelos Moleculares , Peptídeos
4.
Org Biomol Chem ; 14(11): 3120-9, 2016 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-26906931

RESUMO

The substrate scope of fluorinase enzyme mediated transhalogenation reactions is extended. Substrate tolerance allows a peptide cargo to be tethered to a 5'-chloro-5'-deoxynucleoside substrate for transhalogenation by the enzyme to a 5'-fluoro-5'-deoxynucleoside. The reaction is successfully extended from that previously reported for a monomeric cyclic peptide (cRGD) to cargoes of dendritic scaffolds carrying two and four cyclic peptide motifs. The RGD peptide sequence is known to bind upregulated αVß3 integrin motifs on the surface of cancer cells and it is demonstrated that the fluorinated products have a higher affinity to αVß3 integrin than their monomeric counterparts. Extending the strategy to radiolabelling of the peptide cargoes by tagging the peptides with [(18)F]fluoride was only moderately successful due to the poor water solubility of these higher order peptide scaffolds although the strategy holds promise for peptide constructs with improved solubility.


Assuntos
Proteínas de Bactérias/metabolismo , Desoxirribonucleosídeos/química , Desoxirribonucleosídeos/metabolismo , Oxirredutases/metabolismo , Peptídeos Cíclicos/química , Peptídeos Cíclicos/metabolismo , Streptomyces/enzimologia , Desoxirribose/análogos & derivados , Desoxirribose/metabolismo , Halogenação , Humanos , Integrina alfaVbeta3/metabolismo , Modelos Moleculares
5.
Breast Cancer Res Treat ; 144(2): 241-8, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24522376

RESUMO

HER-2 overexpression does not guarantee response to HER2-targeting drugs such as trastuzumab, which is cardiotoxic and expensive, so early detection of response status is crucial. Factors influencing [(18)F]FDG incorporation in the timeframe of cell signalling down-regulation subsequent to trastuzumab treatment are investigated to provide a better understanding of the relationship between growth response and modulation of [(18)F]FDG incorporation. HER-2-overexpressing breast tumour cell lines, MDA-MB-453, SKBr3 and BT474 and MDA-MB-468 (HER2 non-over-expressor) were treated with trastuzumab (4 h) and probed for AKT, pAKT, ERK1/2, pERK1/2 and HIF-1α to determine early signalling pathway inhibitory effects of trastuzumab. Cells incubated with trastuzumab and/or PI3K inhibitor LY294002 and ERK1/2 inhibitor U0126 and glucose transport and [(18)F]FDG incorporation measured. Cell lines expressed AKT, pAKT, ERK1/2 and pERK1/2 but not HIF-1α. Trastuzumab treatment decreased pAkt but not pERK1/2 levels. Trastuzumab did not further inhibit AKT when maximally inhibited with LY294002. Treatment with LY294002 and trastuzumab for 4 h decreased [(18)F]FDG incorporation in BT474 and MDA-MB-453 but not SKBr3 cells. LY294002 inhibited glucose transport by each cell line, but the glucose transport rate was tenfold higher by SKBr3 cells than BT474 and MDA-MB-453 cells. AKT-induced uptake of [(18)F]FDG was found to be HIF-1α independent in breast cancer cell lines. AKT inhibition level and tumour cell glucose transport rate can influence whether or not PI3K inhibitors affect [(18)F]FDG incorporation which may account for the variation in preclinical and clinical findings associated with [(18)F]FDG-PET in response to trastuzumab and other HER-2 targeting drugs.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Neoplasias da Mama/metabolismo , Fluordesoxiglucose F18/farmacocinética , Glucose/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Compostos Radiofarmacêuticos/farmacocinética , Transporte Biológico/efeitos dos fármacos , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Cromonas/farmacologia , Regulação para Baixo/efeitos dos fármacos , Interações Medicamentosas , Feminino , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Morfolinas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Cintilografia , Receptor ErbB-2/metabolismo , Trastuzumab
6.
Org Biomol Chem ; 11(27): 4551-8, 2013 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-23733207

RESUMO

The utility of 5-fluoro-5-deoxyribose (FDR) as an efficient bioconjugation agent for radiolabelling of the RGD peptides c(RGDfK) and c(RGDfC) is demonstrated. The bioconjugation is significantly superior to that achieved with 2-fluoro-2-deoxyglucose (FDG) and benefits from the location of the fluorine at C-5, and that ribose is a 5-membered ring sugar rather than a 6-membered ring. Both features favour ring opening to the aldehydic form of the sugar to promote smooth oxime ligation with aminooxy ether functionalised peptides. [(18)F]FDR was prepared in this study by synthesis from fluoride-18 using an automated synthesis protocol adapting that used routinely for [(18)F]FDG. c(RGDfK) was functionalised with an aminooxyacetyl group (Aoa) via its lysine terminus, while c(RGDfC) was functionalised with an aminooxyhexylmaleimide (Ahm) through a cysteine-maleimide conjugation. Bioconjugation of [(18)F]FDR to c(RGDfC)-Ahm proved to be more efficient than c(RGDfK)-Aoa (92% versus 65%). The unlabelled ((19)F) bioconjugates c(RGDfK)-Aoa-FDR and c(RGDfC)-Ahm-FDR were prepared and their in vitro affinity to purified integrin αvß3 was determined. c(RGDfK)-Aoa-FDR showed the greater affinity. Purified "hot" bioconjugates c(RGDfK)-Aoa-[(18)F]FDR and c(RGDfC)-Ahm-[(18)F]FDR were assayed by incubation with MCF7, LNCaP and PC3 cell lines. In both cases the conjugated RGD peptides showed selectivity for PC3 cells, which express αvß3 integrin, with the c(RGDfK)-Aoa-[(18)F]FDR demonstrating better binding, consistent with its higher in vitro affinity. The study demonstrates that [(18)F]FDR is an efficient bioconjugation ligand for RGD bioactive peptides.


Assuntos
Fluordesoxiglucose F18/química , Integrina alfaVbeta3/análise , Oligopeptídeos/química , Tomografia por Emissão de Pósitrons/métodos , Linhagem Celular Tumoral , Fluordesoxiglucose F18/metabolismo , Humanos , Integrina alfaVbeta3/metabolismo , Oligopeptídeos/metabolismo
7.
Eur Radiol ; 22(7): 1465-78, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22476502

RESUMO

The evaluation of drug pharmacodynamics and early tumour response are integral to current clinical trials of novel cancer therapeutics to explain or predict long term clinical benefit or to confirm dose selection. Tumour vascularity assessment by positron emission tomography could be viewed as a generic pharmacodynamic endpoint or tool for monitoring response to treatment. This review discusses methods for semi-quantitative and quantitative assessment of tumour vascularity. The radioligands and radiotracers range from direct physiological functional tracers like [(15)O]-water to macromolecular probes targeting integrin receptors expressed on neovasculature. Finally we make recommendations on ways to incorporate such measurements of tumour vascularity into early clinical trials of novel therapeutics. Key Points • [ ( 15 ) O]-water is the gold standard for blood flow/tissue perfusion with PET • In some instances dynamic [ ( 18 ) F]-FDG uptake may be used to estimate perfusion • Radiopharmaceuticals that target integrins are now being evaluated for measuring tumour vascularity.


Assuntos
Ensaios Clínicos como Assunto/normas , Neoplasias/diagnóstico por imagem , Neovascularização Patológica/diagnóstico por imagem , Tomografia por Emissão de Pósitrons/normas , Guias de Prática Clínica como Assunto , Compostos Radiofarmacêuticos/normas , Europa (Continente) , Humanos , Neoplasias/irrigação sanguínea , América do Norte , Padrões de Referência
8.
Insights Imaging ; 12(1): 90, 2021 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-34213667

RESUMO

Lung cancer has the highest mortality rate of any tumour type. The main driver of lung tumour growth and development is uncontrolled cellular proliferation. Poor patient outcomes are partly the result of the limited range of effective anti-cancer therapies available and partly due to the limited accuracy of biomarkers to report on cell proliferation rates in patients. Accordingly, accurate methods of diagnosing, staging and assessing response to therapy are crucial to improve patient outcomes. One effective way of assessing cell proliferation is to employ non-invasive evaluation using 3'-deoxy-3'-[18F]fluorothymidine ([18F]FLT) positron emission tomography [18F]FLT-PET. [18F]FLT, unlike the most commonly used PET tracer [18F]fluorodeoxyglucose ([18F]FDG), can specifically report on cell proliferation and does not accumulate in inflammatory cells. Therefore, this radiotracer could exhibit higher specificity in diagnosis and staging, along with more accurate monitoring of therapy response at early stages in the treatment cycle. This review summarises and evaluates published studies on the clinical use of [18F]FLT to diagnose, stage and assess response to therapy in lung cancer.

9.
Ann Clin Transl Neurol ; 7(9): 1580-1593, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32790171

RESUMO

OBJECTIVE: The purpose of the study was to determine the extent and role of systemic hypoxia in the pathogenesis of spinal muscular atrophy (SMA). METHODS: Hypoxia was assayed in vivo in early-symptomatic (postnatal day 5) SMA-model mice by pimonidazole and [18 F]-Fluoroazomycin arabinoside injections, which accumulate in hypoxic cells, followed by immunohistochemistry and tracer biodistribution evaluation. Glucose uptake in hypoxic cells was assayed by [18 F]-Fluorodeoxyglucose labeling. In vitro knockdown of Survival Motor Neuron (SMN) was performed on motor neurons and lactate metabolism measured biochemically, whereas cell cycle progression and cell death were assayed by flow cytometry. RESULTS: All assays found significant levels of hypoxia in multiple organ systems in early symptomatic SMA mouse pups, except aerated tissues such as skin and lungs. This was accompanied by significantly increased glucose uptake in many affected organs, consistent with a metabolic hypoxia response. SMN protein levels were shown to vary widely between motor neuron precursors in vitro, and those with lower levels were most susceptible to cell death. In addition, SMA-model motor neurons were particularly sensitive to hypoxia, with reduced ability to transport lactate out of the cell in hypoxic culture, and a failure in normal cell cycle progression. INTERPRETATION: Not only is there widespread tissue hypoxia and multi-organ cellular hypoxic response in SMA model mice, but SMA-model motor neurons are especially susceptible to that hypoxia. The data support the hypothesis that vascular defects leading to hypoxia are a significant contributor to disease progression in SMA, and offer a route for combinatorial, non-SMN related therapy.


Assuntos
Hipóxia/complicações , Hipóxia/metabolismo , Redes e Vias Metabólicas , Neurônios Motores/metabolismo , Atrofia Muscular Espinal/etiologia , Atrofia Muscular Espinal/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos
10.
Clin Cancer Res ; 14(13): 4326-35, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18594016

RESUMO

PURPOSE: The aims of this study were to investigate whether the cyclin-dependent kinase inhibitor seliciclib could synergize with agents that target ErbB receptors and to elucidate the molecular mechanism of the observed synergy. EXPERIMENTAL DESIGN: Synergy between seliciclib and ErbB receptor targeted agents was investigated in various cell lines using the Calcusyn median effect model. The molecular mechanism of the observed synergy was studied in cultured cells, and the combination of seliciclib and the epidermal growth factor receptor (EGFR) inhibitor erlotinib was evaluated in an H358 xenograft model. RESULTS: Seliciclib synergized with the anti-HER2 antibody trastuzumab in a breast cancer cell line, which overexpresses the HER2 receptor, and with the erlotinib analogue AG1478 in non-small cell lung cancer cell lines. In the H358 non-small cell lung cancer cell line, synergy involved decreased signaling from the EGFR, with AG1478 directly inhibiting kinase activity while seliciclib decreased the levels of key components of the receptor signaling pathway, resulting in enhanced loss of phosphorylated extracellular signal-regulated kinase and cyclin D1. The combination of seliciclib and erlotinib was evaluated further in an H358 xenograft and shown to be significantly more active than either agent alone. An enhanced loss of cyclin D1 was also seen in vivo. CONCLUSIONS: This is the first report that investigates combining seliciclib with an EGFR inhibitor. The combination decreased signaling from the EGFR in vitro and in vivo and was effective in cell lines containing either wild-type or mutant EGFR, suggesting that it may expand the range of tumors that respond to erlotinib, and therefore, such combinations are worth exploring in the clinic.


Assuntos
Antineoplásicos/farmacologia , Regulação Neoplásica da Expressão Gênica , Purinas/farmacologia , Receptor ErbB-2/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Ciclina D1/metabolismo , Sinergismo Farmacológico , Feminino , Humanos , Concentração Inibidora 50 , Camundongos , Modelos Biológicos , Mutação , Transplante de Neoplasias , Receptor ErbB-2/metabolismo , Roscovitina
11.
Cell Death Dis ; 10(11): 861, 2019 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-31723121

RESUMO

The vascular system of the spinal cord is particularly complex and vulnerable. Damage to the main vessels or alterations to the regulation of blood flow will result in a reduction or temporary cessation of blood supply. The resulting tissue hypoxia may be brief: acute, or long lasting: chronic. Damage to the vascular system of the spinal cord will develop after a traumatic event or as a result of pathology. Traumatic events such as road traffic accidents, serious falls and surgical procedures, including aortic cross-clamping, will lead to an immediate cessation of perfusion, the result of which may not be evident for several days, but may have long-term consequences including neurodegeneration. Pathological events such as arterial sclerosis, venous occlusion and spinal cord compression will result in a progressive reduction of blood flow, leading to chronic hypoxia. While in some situations the initial pathology is exclusively vascular, recent research in neurodegenerative disease has drawn attention to concomitant vascular anomalies in disorders, including amyotrophic lateral sclerosis, spinal muscular atrophy and muscular sclerosis. Understanding the role of, and tissue response to, chronic hypoxia is particularly important in these cases, where inherent neural damage exacerbates the vulnerability of the nervous system to stressors including hypoxia.


Assuntos
Hipóxia/metabolismo , Doenças Neurodegenerativas/metabolismo , Fluxo Sanguíneo Regional , Medula Espinal/irrigação sanguínea , Humanos , Hipóxia/patologia , Doenças Neurodegenerativas/patologia , Fluxo Sanguíneo Regional/fisiologia , Medula Espinal/metabolismo , Medula Espinal/patologia , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia
12.
Mol Imaging Biol ; 20(1): 27-36, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28695371

RESUMO

PURPOSE: Molecular imaging of αvß3 integrin has exhibited real potential to guide the appropriate use of anti-angiogenic therapies. However, an incomplete understanding of the factors that influence binding of αvß3 integrin-specific radiotracers currently limits their use for assessing response to therapy in cancer patients. This study identifies two fundamental factors that modulate uptake of these radiotracers. Procedures Experiments were performed in prostate cancer (PC3) and glioblastoma (U87MG) cells, which differentially express αvß3 integrin. αvß3 integrin-specific radiotracers were used to investigate the effect of manipulating αvß3 integrin expression or activation in cellular binding assays. ß3 integrin and αvß3 integrin expression were measured by western blotting and flow cytometry, respectively. The effect of select pharmacological inhibitors on αvß3 integrin activation and expression was also determined. RESULTS: Radiotracer binding was proportional to αvß3 integrin expression when it was decreased (ß3 knock-down cells) or increased, either using pharmacological inhibitors of cell signalling or by culturing cells for different times. Studies with both small molecule and arginine-glycine-aspartic acid (RGD)-based radiotracers revealed increased radiotracer binding after activation of αvß3 integrin with Mn2+ or talin head domain. Moreover, inhibition of fundamental signalling pathways (mitogen-activated protein kinase kinase (MEK), Src and VEGFR2) decreased radiotracer binding, reflecting reduced αvß3 integrin activity. CONCLUSION: Binding of small molecule ligands and radiolabelled RGD peptides is modulated by expression and activation status of αvß3 integrin. αvß3 integrin-specific radiotracers can provide otherwise inaccessible information of the effect of signalling pathways on αvß3 integrin. This has significant implications for assessing response to anti-angiogenic therapies in clinical studies.


Assuntos
Integrina alfaVbeta3/metabolismo , Compostos Radiofarmacêuticos/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Ligação Proteica , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
13.
Hypoxia (Auckl) ; 5: 45-59, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28580362

RESUMO

PURPOSE: There is an urgent need to develop effective therapies and treatment strategies to treat hypoxic tumors, which have a very poor prognosis and do not respond well to existing therapies. METHODS: A novel hypoxia-targeting agent, KEMTUB012-NI2, was synthesized by conjugating a 2-nitroimidazole hypoxia-targeting moiety to a synthetic tubulysin, a very potent antimitotic. Its hypoxic selectivity and mode of action were studied in breast cancer cell lines. RESULTS: KEMTUB012-NI2 exhibited a similar selectivity for hypoxic cells to that of tirapazamine, a well-established hypoxia-targeting agent, but was >1,000 times more potent in cell cytotoxicity assays. The hypoxia-targeting mechanism for both KEMTUB012-NI2 and tirapazamine was selective and mediated by one-electron reductases. However, while cytochrome p450 reductase (POR) downregulation could inhibit tirapazamine cytotoxicity, it actually sensitized hypoxic cells to KEMTUB012-NI2. CONCLUSION: KEMTUB012-NI2 is a potent new agent that can selectively target hypoxic cancer cells. The hypoxia selectivity of KEMTUB012-NI2 and tirapazamine appears to be differentially activated by reductases. Since reductases are heterogeneously expressed in tumors, the different activation mechanisms will allow these agents to complement each other. Combining POR downregulation with KEMTUB012-NI2 treatment could be a new treatment strategy that maximizes efficacy toward hypoxic tumor cells while limiting systemic toxicity.

14.
ChemMedChem ; 12(14): 1142-1151, 2017 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-28608961

RESUMO

Nonpeptidic Arg-Gly-Asp (RGD)-mimic ligands were designed and synthesized by click chemistry between an arginine-azide mimic and an aspartic acid-alkyne mimic. Some of these molecules combine excellent in vitro properties (high αv ß3 affinity, selectivity, drug-like logD, high metabolic stability) with a variety of radiolabeling options (e.g., tritium and fluorine-18, plus compatibility with radio-iodination), not requiring the use of chelators or prosthetic groups. The binding mode of the resulting triazole RGD mimics to αv ß3 or αIIb ß3 receptors was investigated by molecular modeling simulations. Lead compound 12 was successfully radiofluorinated and used for in vivo positron emission tomography/computed tomography (PET/CT) studies in U87 tumor models, which showed only modest tumor uptake and retention, owing to rapid excretion. These results demonstrate that the novel click RGD mimics are excellent radiolabeled probes for in vitro and cell-based studies on αv ß3 integrin, whereas further optimization of their pharmacokinetic and dynamic profiles is necessary for successful use in in vivo imaging.


Assuntos
Integrina alfaVbeta3/química , Oligopeptídeos/síntese química , Peptidomiméticos/síntese química , Linhagem Celular Tumoral , Química Click , Radioisótopos de Flúor , Humanos , Integrina alfaVbeta3/metabolismo , Radioisótopos do Iodo , Modelos Moleculares , Imagem Molecular , Oligopeptídeos/química , Peptidomiméticos/química , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/química , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Relação Estrutura-Atividade , Trítio
15.
Sci Rep ; 6: 36544, 2016 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-27811956

RESUMO

Akt is an intracellular signalling pathway that serves as an essential link between cell surface receptors and cellular processes including proliferation, development and survival. The pathway has many downstream targets including glycogen synthase kinase3 which is a major regulatory kinase for cell cycle transit as well as controlling glycogen synthase activity. The Akt pathway is frequently up-regulated in cancer due to overexpression of receptors such as the epidermal growth factor receptor, or mutation of signalling pathway kinases resulting in inappropriate survival and proliferation. Consequently anticancer drugs have been developed that target this pathway. MDA-MB-468 breast and HCT8 colorectal cancer cells were treated with inhibitors including LY294002, MK2206, rapamycin, AZD8055 targeting key kinases in/associated with Akt pathway and the consistency of changes in 31P-NMR-detecatable metabolite content of tumour cells was examined. Treatment with the Akt inhibitor MK2206 reduced phosphocholine levels in MDA-MB-468 cells. Treatment with either the phosphoinositide-3-kinase inhibitor, LY294002 and pan-mTOR inhibitor, AZD8055 but not pan-Akt inhibitor MK2206 increased uridine-5'-diphosphate-hexose cell content which was suppressed by co-treatment with glycogen synthase kinase 3 inhibitor SB216763. This suggests that there is an Akt-independent link between phosphoinositol-3-kinase and glycogen synthase kinase3 and demonstrates the potential of 31P-NMR to probe intracellular signalling pathways.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cromonas/farmacologia , Receptores ErbB/metabolismo , Compostos Heterocíclicos com 3 Anéis/farmacologia , Humanos , Espectroscopia de Ressonância Magnética/métodos , Morfolinas/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR/antagonistas & inibidores
16.
Biochem J ; 379(Pt 3): 641-51, 2004 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-14769130

RESUMO

In 1321N1 astrocytoma cells, stimulation of the IGF-1 (insulin-like growth factor-1) receptor increased the association of PI3K [phosphoinositide (PI) 3-kinase] activity with IRS-1 (insulin re-ceptor substrate 1), and increased the cellular concentration of PtdIns(3,4,5)P3. Carbachol, acting on M3 muscarinic receptors, inhibited insulin-, but not PDGF (platelet-derived growth factor)-, stimulated responses by approximately 50%. The inhibition of IRS-1-associated PI3K activity by carbachol (i) was rapid (<1 min), persistent (> or =60 min) and potent (half-maximal concentration approximately 1 microM); (ii) was reproduced by stimuli for several phospholipase-C-coupled receptors; (iii) was prevented by the inhibition of protein kinase C, but not by chelation of intracellular Ca2+; and (iv) was not blocked or reproduced by inhibitors or stimuli respectively of mitogen-activated protein kinase, PI3K, protein kinase B or the mammalian target of rapamycin. However, the effects of carbachol were prevented by sodium vanadate, a protein tyrosine phosphatase inhibitor, and were accompanied by reduced insulin-stimulated IRS-1 tyrosine phosphorylation and recruitment of the 85 kDa regulatory subunit of PI3K to IRS-1, but not by reduced IGF-1 receptor kinase activity. The inhibitory effect of carbachol was reproduced by okadaic acid, a protein serine/threonine phosphatase inhibitor, but not by PDGF, yet all three agents stimulated the serine phosphorylation of IRS-1 at residues Ser312, Ser616 and Ser636/639, albeit to different extents. Thus muscarinic receptors may inhibit insulin signalling by promoting IRS-1 tyrosine dephosphorylation and/or by uncoupling IRS-1 from the stimulated IGF-1 receptor by stimulating IRS-1 serine phosphorylation. However, the proportion of IRS-1 molecules phosphorylated at a particular site or the phosphorylation of additional IRS-1 serine residues other than those noted above must be important.


Assuntos
Astrocitoma/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Receptor IGF Tipo 1/antagonistas & inibidores , Receptores Muscarínicos/metabolismo , Transdução de Sinais , Astrocitoma/patologia , Cálcio/metabolismo , Carbacol/farmacologia , Linhagem Celular Tumoral , Ativação Enzimática/efeitos dos fármacos , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Insulina/farmacologia , Proteínas Substratos do Receptor de Insulina , Ionomicina/farmacologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Ácido Okadáico/farmacologia , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Fosfosserina/metabolismo , Fosfotirosina/metabolismo , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteína Quinase C/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Receptor IGF Tipo 1/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais/efeitos dos fármacos , Acetato de Tetradecanoilforbol/farmacologia , Fosfolipases Tipo C/metabolismo
17.
Biochem J ; 377(Pt 3): 653-63, 2004 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-14604433

RESUMO

PtdIns(3,4) P (2), a breakdown product of the lipid second messenger PtdIns(3,4,5) P (3), is a key signalling molecule in pathways controlling various cellular events. Cellular levels of PtdIns(3,4) P (2) are elevated upon agonist stimulation, mediating downstream signalling pathways by recruiting proteins containing specialized lipid-binding modules, such as the pleckstrin homology (PH) domain. A recently identified protein, TAPP1 (tandem-PH-domain-containing protein 1), has been shown to interact in vitro with high affinity and specificity with PtdIns(3,4) P (2) through its C-terminal PH domain. In the present study, we have utilized this PH domain tagged with glutathione S-transferase (GST-TAPP1-PH) as a probe in an on-section immunoelectron microscopy labelling procedure, mapping the subcellular distribution of PtdIns(3,4) P (2). As expected, we found accumulation of PtdIns(3,4) P (2) at the plasma membrane in response to the agonists platelet-derived growth factor and hydrogen peroxide. Importantly, however, we also found agonist stimulated PtdIns(3,4) P (2) labelling of intracellular organelles, including the endoplasmic reticulum and multivesicular endosomes. Expression of the 3-phosphatase PTEN (phosphatase and tensin homologue deleted on chromosome 10) in PTEN-null U87MG cells revealed differential sensitivity of these lipid pools to the enzyme. These data suggest a role for PtdIns(3,4) P (2) in endomembrane function.


Assuntos
Proteínas Sanguíneas/genética , Proteínas de Transporte/genética , Peptídeos e Proteínas de Sinalização Intracelular , Espaço Intracelular/metabolismo , Proteínas de Membrana/genética , Fosfatos de Fosfatidilinositol/metabolismo , Fosfoproteínas/genética , Homologia de Sequência do Ácido Nucleico , Animais , Linhagem Celular , Linhagem Celular Tumoral , Sondas de DNA/genética , Regulação para Baixo/efeitos dos fármacos , Retículo Endoplasmático/química , Retículo Endoplasmático/efeitos dos fármacos , Humanos , Peróxido de Hidrogênio/farmacologia , Membranas Intracelulares/química , Membranas Intracelulares/efeitos dos fármacos , Espaço Intracelular/química , Camundongos , Microscopia Imunoeletrônica/métodos , PTEN Fosfo-Hidrolase , Peptídeos/genética , Monoéster Fosfórico Hidrolases/biossíntese , Fator de Crescimento Derivado de Plaquetas/farmacologia , Estrutura Terciária de Proteína/genética , Coloração e Rotulagem/métodos , Células Swiss 3T3/química , Células Swiss 3T3/efeitos dos fármacos , Proteínas Supressoras de Tumor/biossíntese
18.
Biochem J ; 382(Pt 3): 857-65, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15242348

RESUMO

Binding of the Rac1-specific guanine-nucleotide-exchange factor, Tiam1, to the plasma membrane requires the N-terminal pleckstrin homology domain. In the present study, we show that membrane-association is mediated by binding of PtdIns(4,5)P(2) to the pleckstrin homology domain. Moreover, in 1321N1 astrocytoma cells, translocation of Tiam1 to the cytosol, following receptor-mediated stimulation of PtdIns(4,5)P(2) breakdown, correlates with decreased Rac1-GTP levels, indicating that membrane-association is required for GDP/GTP exchange on Rac1. In addition, we show that platelet-derived growth factor activates Rac1 in vivo by increasing PtdIns(3,4,5)P(3) concentrations, rather than the closely related lipid, PtdIns(3,4)P(2). Finally, the data demonstrate that PtdIns(4,5)P(2) and PtdIns(3,4,5)P(3) bind to the same pleckstrin homology domain in Tiam1 and that soluble inositol phosphates appear to compete with lipids for this binding. Together, these novel observations provide strong evidence that distinct phosphoinositides regulate different functions of this enzyme, indicating that local concentrations of signalling lipids and the levels of cytosolic inositol phosphates will play crucial roles in determining its activity in vivo.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Androstadienos/farmacologia , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Citosol/metabolismo , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Fragmentos de Peptídeos/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatos de Fosfatidilinositol/fisiologia , Inibidores de Fosfoinositídeo-3 Quinase , Fator de Crescimento Derivado de Plaquetas/farmacologia , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteína 1 Indutora de Invasão e Metástase de Linfoma de Células T , Trombina/metabolismo , Wortmanina
19.
J Cancer Res Clin Oncol ; 141(9): 1523-32, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25579456

RESUMO

PURPOSES: Metformin, currently undergoing clinical trials as an adjuvant for the treatment of breast cancer, modulates the activity of key intracellular signalling molecules which affect 2-[(18)F]Fluoro-2-deoxy-D-glucose ([(18)F]FDG) incorporation. Here, we investigate the effect of drugs used in the treatment of breast cancer combined with metformin on [(18)F]FDG incorporation in HER2- or HER1-overexpressing breast cancer cells to determine whether or not metformin may obscure changes in [(18)F]FDG incorporation induced by clinically utilised anticancer drugs in the treatment of breast cancer. METHODS: Three breast cancer cell lines expressing HER2 and one HER2 negative but HER1 positive were exposed to metformin, doxorubicin and trastuzumab or cetuximab. Cytotoxicity was measured by the MTT assay. Expression of active (phospho-) AMPK, PKB (Akt) and ERK was determined by Western blotting. [(18)F]FDG incorporation by cells exposed to drug combinations with metformin was determined. Glucose transport was assessed by measuring the initial rate of uptake of [(3)H]O-methyl-D-glucose ([(3)H]OMG). Phosphorylation of [(18)F]FDG was determined in intact cells after exposure to [(18)F]FDG. RESULTS: Phospho-AMPK was increased by metformin in all cell lines whilst phospho-Akt and phospho-ERK expressions were decreased in two. Metformin treatment increased [(18)F]FDG incorporation in all cell lines, and treatment with anti-HER antibodies or doxorubicin only produced minor modulations in the increase induced by metformin alone. Glucose transport was increased in BT474 cells and decreased in SKBr3 and MDA-MB-468 cells after treatment with metformin. The fraction of phosphorylated [(18)F]FDG was increased in metformin-treated cells compared with controls, suggesting that hexokinase efficiency was increased by metformin. CONCLUSION: This is the first study to show that increased [(18)F]FDG incorporation by breast cancer cells induced by metformin overwhelms the effect of doxorubicin and anti-HER treatments on [(18)F]FDG incorporation. Metformin-induced increased [(18)F]FDG incorporation was consistently associated with enhanced [(18)F]FDG phosphorylation.


Assuntos
Anticorpos Monoclonais/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Doxorrubicina/farmacologia , Fluordesoxiglucose F18/farmacocinética , Metformina/farmacologia , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Neoplasias da Mama/diagnóstico por imagem , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular Tumoral , Cetuximab/administração & dosagem , Cetuximab/farmacologia , Doxorrubicina/administração & dosagem , Interações Medicamentosas , Receptores ErbB/imunologia , Feminino , Humanos , Metformina/administração & dosagem , Cintilografia , Compostos Radiofarmacêuticos/farmacocinética , Transdução de Sinais/efeitos dos fármacos , Trastuzumab/administração & dosagem , Trastuzumab/farmacologia
20.
J Cancer Res Clin Oncol ; 141(9): 1575-83, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25633717

RESUMO

PURPOSE: Tubulysins are natural tetrapeptides that inhibit tubulin polymerisation. Tubulysins are very potent inhibitors of mammalian cancer cell growth, but restricted availability has limited their characterisation and development as anti-cancer compounds. KEMTUB10 was recently developed as a synthetic analogue of natural tubulysins. METHODS: The cell cytotoxicity of KEMTUB10 was studied in cell lines that represent the main breast cancer sub-types. The KEMTUB10 pro-apoptotic mechanism of action was studied in MCF7 and MDAMB231 cells. RESULTS: KEMTUB10 exerts a potent cytotoxic effect in cells representing the main breast cancer sub-types. KEMTUB10 blocks cells in the G2/M phase of the cell cycle and is a strong stimulator of apoptosis/cell death. KEMTUB10-induced apoptosis involves p53 and Bim, and to some extent Bcl-2 phosphorylation. CONCLUSIONS: KEMTUB10 is a promising new anti-mitotic that exerts a potent cytotoxic effect in breast cancer cells, blocks cells in the G2/M phase of the cell cycle and stimulates apoptosis/cell death. KEMTUB10 has a distinct mode of action to taxol, appears to be sensitive to different molecular factors in cells and is likely subject to different mechanisms of acquired resistance. KEMTUB10 has the potential to be an important addition to the anti-cancer therapeutic armoury.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Proteínas de Membrana/metabolismo , Oligopeptídeos/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteína 11 Semelhante a Bcl-2 , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Células MCF-7 , Receptor ErbB-2/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA