Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Dalton Trans ; 53(13): 6031-6040, 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38470348

RESUMO

BOLD-100 (formerly IT-139, KP1339), a well-established chemotherapeutic agent, is currently being investigated in clinical trials for the treatment of gastric, pancreatic, colorectal, and bile duct cancer. Despite numerous studies, the exact mode of action is still the subject of discussions. Radiolabeled BOLD-100 could be a powerful tool to clarify pharmacokinetic pathways of the compound and to predict therapy responses in patients using nuclear molecular imaging prior to the therapy. In this study, the radiosyntheses of carrier-added (c.a.) [97/103Ru]BOLD-100 were performed with the two ruthenium isotopes ruthenium-103 (103Ru; ß-, γ) and ruthenium-97 (97Ru; EC, γ), of which in particular the latter isotope is suitable for imaging by single-photon emission computed tomography (SPECT). To identify the best tumor-to-background ratio for diagnostic imaging, biodistribution studies were performed with two different injected doses of c.a. [103Ru]BOLD-100 (3 and 30 mg kg-1) in Balb/c mice bearing CT26 allografts over a time period of 72 h. Additionally, ex vivo autoradiography of the tumors (24 h p.i.) was conducted. Our results indicate that the higher injected dose (30 mg kg-1) leads to more unspecific accumulation of the compound in non-targeted tissue, which is likely due to an overload of the albumin transport system. It was also shown that lower amounts of injected c.a. [103Ru]BOLD-100 resulted in a relatively higher tumor uptake and, therefore, a better tumor-to-background ratio, which are encouraging results for future imaging studies using c.a. [97Ru]BOLD-100.


Assuntos
Antineoplásicos , Neoplasias , Compostos Organometálicos , Radioisótopos de Rutênio , Rutênio , Animais , Camundongos , Humanos , Distribuição Tecidual , Antineoplásicos/farmacologia
2.
Curr Pharm Des ; 29(13): 1067, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37231765

RESUMO

An article was published in the journal "Current Pharmaceutical Design", Volume 9, No. 25, 2003, pp: 2078-2089 [1]. The first author is requesting an alteration in the name. Details of a correction are provided here. The original name published was Markus Galanski. The request is to change the name to Mathea Sophia Galanski. The original article can be found online at: https://www.eurekaselect.com/article/8545 We regret the error and apologize to readers.

3.
J Dairy Sci ; 104(7): 7711-7724, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33896629

RESUMO

Lifting the preweaning milk restriction in dairy calves has been causally associated with beneficial effects on growth and future lactation performance. However, the biological mechanisms linking early-life nutrient supply and future performance remain insufficiently understood. Thus, the objective of this study was to characterize growth and the metabolic profiles of calves fed a restricted (RES) and an elevated (ELE) milk supply preweaning. A total of 86 female Holstein Friesian calves were blocked in pairs by maternal parity and received identical colostrum supply within block. Treatments randomized within block consisted of a milk replacer (MR; 24% crude protein, 18% crude fat, and 45% lactose) supplied at either 5.41 Mcal of ME in 8 L of MR/d (ELE) or 2.71 Mcal of ME in 4 L of MR/d (RES) from d 2 after birth until they were stepped down by 50% during wk 7 and fully weaned at wk 8. All calves had ad libitum access to pelleted calf starter (17.3% crude protein, 24.4% neutral detergent fiber, 2.0% crude fat, and 18.2% starch), chopped wheat straw, and water. At 2 and 49 d of age, blood samples were taken for metabolomics analysis. The ELE group by design consumed more milk replacer, resulting in a lower starter intake and a greater body weight and average daily gain. The ELE calves consumed 20.7% more ME and 9.7% more crude protein. However, efficiency of growth was not different between groups. Metabolomic profiling using 908 identified metabolites served to characterize treatment-dependent biochemical differences. Principal component analysis revealed clearly distinct metabolic profiles at 49 d of age in response to preweaning milk supply. Changes in energy (fatty acid metabolism and tricarboxylic acid metabolites), protein (free AA, dipeptides, and urea cycle), and liver metabolism (bile acid and heme metabolism) were the main effects associated with the dietary differences. The ELE group consumed proportionately more glucogenic nutrients via milk replacer, whereas the RES group consumed proportionately more ketogenic nutrients from the digestion of the calf starter, comprising a larger portion of total intake. Associated with the higher growth rate of the ELE group, hepatic changes were expressed as differences in bile acid and heme metabolism. Furthermore, energy metabolism differences were noted in fatty acid and AA metabolism and the urea cycle. The metabolic profile differences between the ELE and RES groups reflect the broad differences in nutrient intake and diet composition and might point to which metabolic processes are responsible for greater dairy performance for cows fed a greater milk supply preweaning.


Assuntos
Ração Animal , Substitutos do Leite , Ração Animal/análise , Animais , Peso Corporal , Bovinos , Dieta/veterinária , Metabolismo Energético , Feminino , Metabolômica , Leite , Nutrientes , Gravidez , Desmame
4.
Analyst ; 143(9): 1997-2001, 2018 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-29629473

RESUMO

Pt(iv) prodrugs are a class of promising anticancer agents, which are activated by reduction to the active Pt(ii) species. Consequently, the reduction process is a crucial parameter. Herein, a new approach using electrochemistry (EC) coupled to liquid chromatography (LC) and electrospray ionization-mass spectrometry (ESI-MS) or inductively coupled plasma (ICP)-MS was applied. This enabled getting insights into the differences in the reduction and ligand release of platinum(iv) complexes with varying equatorial core structures.

5.
Br J Cancer ; 116(4): 489-500, 2017 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-28095394

RESUMO

BACKGROUND: Colorectal carcinoma (CRC) is the third most common cancer worldwide. Platinum-based anticancer compounds still constitute one mainstay of systemic CRC treatment despite limitations due to adverse effects and resistance development. Trabectedin has shown promising antitumor effects in CRC, however, again resistance development may occur. In this study, we aimed to develop strategies to circumvent or even exploit acquired trabectedin resistance in novel CRC treatment regimens. METHODS: Human HCT116 CRC cells were selected for acquired trabectedin resistance in vitro and characterised by cell biological as well as bioinformatic approaches. In vivo xenograft experiments were conducted. RESULTS: Selection of HCT116 cells for trabectedin resistance resulted in p53-independent hypersensitivity of the selected subline against cisplatin. Bioinformatic analyses of mRNA microarray data suggested deregulation of nucleotide excision repair and particularly loss of the ubiquitin ligase CUL4A in trabectedin-selected cells. Indeed, transient knockdown of CUL4A sensitised parental HCT116 cells towards cisplatin. Trabectedin selected but not parental HCT116 xenografts were significantly responsive towards cisplatin treatment. CONCLUSIONS: Trabectedin selection-mediated CUL4A loss generates an Achilles heel in CRC cancer cells enabling effective cisplatin treatment. Hence, inclusion of trabectedin in cisplatin-containing cancer treatment regimens might cause profound synergism based on reciprocal resistance prevention.


Assuntos
Cisplatino/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Proteínas Culina/genética , Dioxóis/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Tetra-Hidroisoquinolinas/uso terapêutico , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Proteínas Culina/antagonistas & inibidores , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Genes p53 , Células HCT116 , Humanos , RNA Interferente Pequeno/farmacologia , Trabectedina
6.
Dalton Trans ; 46(2): 329-332, 2017 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-27918050

RESUMO

Supramolecular Pt(ii) quadrangular boxes bind native and G-quadruplex DNA motifs in a size-dependent fashion. Three Pt molecular squares of distinct size show biological activity against cancer cells and heavily influence the expression of genes known to form G-quadruplexes in their promoter regions. The smallest Pt-box displays less activity but more selectivity for a quadruplex formed in the c-Kit gene.

7.
Chem Sci ; 7(5): 3052-3061, 2016 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-29997796

RESUMO

Nano-scale secondary ion mass spectrometry (NanoSIMS) enables trace element and isotope analyses with high spatial resolution. This unique capability has recently been exploited in several studies analyzing the subcellular distribution of Au and Pt anticancer compounds. However, these studies were restricted to cell culture systems. To explore the applicability to the in vivo setting, we developed a combined imaging approach consisting of laser ablation inductively coupled plasma mass spectrometry (LA-ICP-MS), NanoSIMS and transmission electron microscopy (TEM) suitable for multi-scale detection of the platinum distribution in tissues. Applying this approach to kidney and tumor samples upon administration of selected platinum(iv) anticancer prodrugs revealed uneven platinum distributions on both the organ and subcellular scales. Spatial platinum accumulation patterns were quantitatively assessed by LA-ICP-MS in histologically heterogeneous organs (e.g., higher platinum accumulation in kidney cortex than in medulla) and used to select regions of interest for subcellular-scale imaging with NanoSIMS. These analyses revealed cytoplasmic sulfur-rich organelles accumulating platinum in both kidney and malignant cells. Those in the tumor were subsequently identified as organelles of lysosomal origin, demonstrating the potential of the combinatorial approach for investigating therapeutically relevant drug concentrations on a submicrometer scale.

8.
Cancer Lett ; 361(1): 112-20, 2015 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-25749419

RESUMO

Although triapine is promising for treatment of advanced leukemia, it failed against solid tumors due to widely unknown reasons. To address this issue, a new triapine-resistant cell line (SW480/tria) was generated by drug selection and investigated in this study. Notably, SW480/tria cells displayed broad cross-resistance against several known ABCB1 substrates due to high ABCB1 levels (induced by promoter hypomethylation). However, ABCB1 inhibition did not re-sensitize SW480/tria cells to triapine and subsequent analysis revealed that triapine is only a weak ABCB1 substrate without significant interaction with the ABCB1 transport function. Interestingly, in chemo-naive, parental SW480 cells short-time (24 h) treatment with triapine stimulated ABCB1 expression. These effects were based on activation of protein kinase C (PKC), a known response to cellular stress. In accordance, SW480/tria cells were characterized by elevated levels of PKC. Together, this led to the conclusion that increased ABCB1 expression is not the major mechanism of triapine resistance in SW480/tria cells. In contrast, increased ABCB1 expression was found to be a consequence of triapine stress-induced PKC activation. These data are especially of importance when considering the choice of chemotherapeutics for combination with triapine.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Proteína Quinase C/metabolismo , Piridinas/farmacologia , Tiossemicarbazonas/farmacologia , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , Hibridização Genômica Comparativa , Metilação de DNA/efeitos dos fármacos , Humanos , Regiões Promotoras Genéticas/genética , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , RNA Mensageiro/genética , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
9.
Dalton Trans ; 44(2): 659-68, 2015 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-25385191

RESUMO

The very promising results of Na-trans-[RuCl4(1H-indazole)2] (NKP-1339) in clinical studies have fuelled renewed interest in the research and development of ruthenium(III) coordination compounds for cancer therapy. By applying an improved synthetic approach to this class of coordination compounds, six new examples of the general formula (cation)-trans-[RuCl4(azole)2], where (cation) = tetrabutylammonium (Bu4N)(+) (1, 2), sodium (3, 4), azolium (5, 6), and azole = 1-methyl-indazole (1, 3, 5), 1-ethyl-indazole (2, 4, 6), have been prepared. All compounds have been characterized by elemental analysis, electrospray ionization (ESI) mass spectrometry, UV-vis-, and NMR-spectroscopy and, if possible, X-ray diffraction analysis. Furthermore, the influence of the alkyl substituent at the position N1 of the indazole backbone on the stability in aqueous media as well as on the biological activity in three human cancer cell lines (CH1, A549, and SW480) and on the cellular accumulation in SW480 cells is discussed.


Assuntos
Antineoplásicos/síntese química , Complexos de Coordenação/farmacologia , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Cristalografia por Raios X , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Espectroscopia de Prótons por Ressonância Magnética , Espectrometria de Massas por Ionização por Electrospray , Análise Espectral/métodos
10.
J Biomed Nanotechnol ; 10(5): 877-84, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24734541

RESUMO

Ruthenium anticancer drugs belong to the most promising non-platinum anticancer metal compounds in clinical evaluation. However, although the clinical results are promising regarding both activity and very low adverse effects, the clinical application is currently hampered by the limited solubility and stability of the drug in aqueous solution. Here, we present a new nanoparticle formulation based on polymer-based micelles loaded with the anticancer lead ruthenium compound KP1019. Nanoprepared KP1019 was characterised by enhanced stability in aqueous solutions. Moreover, the nanoparticle formulation facilitated cellular accumulation of KP1019 (determined by ICP-MS measurements) resulting in significantly lowered IC50 values. With regard to the mode of action, increased cell cycle arrest in G2/M phase (PI-staining), DNA damage (Comet assay) as well as enhanced levels of apoptotic cell death (caspase 7 and PARP cleavage) were found in HCT116 cells treated with the new nanoformulation of KP1019. Summarizing, we present for the first time evidence that nanoformulation is a feasible strategy for improving the stability as well as activity of experimental anticancer ruthenium compounds.


Assuntos
Indazóis/administração & dosagem , Indazóis/química , Nanocápsulas/administração & dosagem , Nanocápsulas/química , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/patologia , Compostos Organometálicos/administração & dosagem , Compostos Organometálicos/química , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Linhagem Celular Tumoral , Difusão , Composição de Medicamentos/métodos , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Humanos , Nanocápsulas/ultraestrutura , Compostos de Rutênio , Resultado do Tratamento
11.
Histochem Cell Biol ; 133(4): 375-404, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20174817

RESUMO

To develop a platform for molecular magnetic resonance imaging, we prepared gadolinium-bearing albumin-polylactic acid nanoparticles in the size range 20-40 nm diameter. Iterative cycles of design and testing upscaled the synthesis procedures to gram amounts for physicochemical characterisation and for pharmacokinetic testing. Morphological analyses showed that the nanoparticles were spheroidal with rough surfaces. Particle sizes were measured by direct transmission electron microscopical measurements from negatively contrasted preparations, and by use of photon correlation spectroscopy; the two methods each documented nanoparticle sizes less than 100 nm and generally 10-40 nm diameter, though with significant intrabatch and interbatch variability. The particles' charge sufficed to hold them in suspension. HSA retained its tertiary structure in the particles. The nanoparticles were stable against turbulent flow conditions and against heat, though not against detergents. MRI imaging of liquid columns was possible at nanoparticle concentrations below 10 mg/ml. The particles were non-cytotoxic, non-thrombogenic and non-immunogenic in a range of assay systems developed for toxicity testing of nanoparticles. They were micellar prior to lyophilisation, but loosely structured aggregated masses after lyophilisation and subsequent resuspension. These nanoparticles provide a platform for further development, based on non-toxic materials of low immunogenicity already in clinical use, not expensive, and synthesized using methods which can be upscaled for industrial production.


Assuntos
Meios de Contraste/química , Gadolínio/química , Espectroscopia de Ressonância Magnética , Nanopartículas/química , Nanopartículas/ultraestrutura , Albuminas/química , Albuminas/ultraestrutura , Imageamento por Ressonância Magnética/métodos , Microscopia Eletrônica de Transmissão , Tamanho da Partícula
12.
Met Based Drugs ; 2009: 681270, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19789639

RESUMO

The effects of indazolium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (KP1019, or FFC14A), the second ruthenium compound that entered clinical trials, in an in vitro model of tumour invasion and metastasis show that the antitumour effects of this compound might include also the modulation of cell behaviour although its cytotoxicity appears to be predominant over these effects. The comparison with its imidazole analogue KP418 shows however its superiority, being able to control in vitro cell growth and in some instances also in vivo tumour development. These results suggest that the activity of KP1019 is predominantly due to direct cytotoxic effects for tumour cells, evident also in vivo on primary tumour growth and that the effects on modulation of the biological behaviour of the cancer cell can be present but might have only a partial role.

13.
Curr Cancer Drug Targets ; 9(5): 595-607, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19508176

RESUMO

KP772 is a new lanthanum complex containing three 1,10-phenathroline molecules. Recently, we have demonstrated that the promising in vitro and in vivo anticancer properties of KP772 are based on p53-independent G(0)G(1) arrest and apoptosis induction. A National Cancer Institute (NCI) screen revealed significant correlation of KP772 activity with that of the ribonucleotide reductase (RR) inhibitor hydroxyurea (HU). Consequently, this study aimed to investigate whether KP772 targets DNA synthesis in tumor cells by RR inhibition. Indeed, KP772 treatment led to significant reduction of cytidine incorporation paralleled by a decrease of deoxynucleoside triphosphate (dNTP) pools. This strongly indicates disruption of RR activity. Moreover, KP772 protected against oxidative stress, suggesting that this drug might interfere with RR by interaction with the tyrosyl radical in subunit R2. Additionally, several observations (e.g. increase of transferrin receptor expression and protective effect of iron preloading) indicate that KP772 interferes with cellular iron homeostasis. Accordingly, co-incubation of Fe(II) with KP772 led to generation of a coloured iron complex (Fe-KP772) in cell free systems. In electron paramagnetic resonance (EPR) measurements of mouse R2 subunits, KP772 disrupted the tyrosyl radical while Fe-KP772 had no significant effects. Moreover, coincubation of KP772 with iron-loaded R2 led to formation of Fe-KP772 suggesting chelation of RR-bound Fe(II). Summarizing, our data prove that KP772 inhibits RR by targeting the iron centre of the R2 subunit. As also Fe-KP772 as well as free lanthanum exert significant -though less pronounced- cytotoxic/static activities, additional mechanisms are likely to synergise with RR inhibition in the promising anticancer activity of KP772.


Assuntos
Antineoplásicos/farmacologia , Fosfatos de Dinucleosídeos/metabolismo , Compostos Organometálicos/farmacologia , Fenantrolinas/farmacologia , Ribonucleotídeo Redutases/antagonistas & inibidores , Linhagem Celular Tumoral , DNA/biossíntese , Sinergismo Farmacológico , Feminino , Humanos , Hidroxiureia/farmacologia , Ferro/metabolismo , Quelantes de Ferro/farmacologia , Nucleotídeos/metabolismo , Espécies Reativas de Oxigênio/antagonistas & inibidores , Receptores da Transferrina/biossíntese
14.
Biochem Pharmacol ; 73(12): 1873-86, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17445775

RESUMO

Recently, we have introduced [tris(1,10-phenanthroline)lanthanum(III)] trithiocyanate (KP772, FFC24) as a new lanthanum compound which has promising anticancer properties in vivo and in vitro. Aim of this study was to investigate the impact of ABC transporter-mediated multidrug resistance (MDR) on the anticancer activity of KP772. Here, we demonstrate that all MDR cell models investigated, overexpressing ABCB1 (P-glycoprotein), ABCC1 (multidrug resistance protein 1), or ABCG2 (breast cancer resistance protein) either due to drug selection or gene transfection, were significantly hypersensitive against KP772. Using ABCB1-overexpressing KBC-1 cells as MDR model, KP772 hypersensitivity was demonstrated to be based on stronger apoptosis induction and/or cell cycle arrest at unaltered cellular drug accumulation. KP772 did neither stimulate ABCB1 ATPase activity nor alter rhodamine 123 accumulation arguing against a direct interaction with ABCB1. Accordingly, several drug resistance modulators did not sensitize but rather protect MDR cells against KP772-induced cytotoxicity. Moreover, long-term KP772 treatment of KBC-1 cells at subtoxic concentrations led within 20 passages to a complete loss of drug resistance based on blocked MDR1 gene expression. When exposing parental KB-3-1 cells to subtoxic, stepwise increasing KP772 concentrations, we observed, in contrast to several other metallo-drugs, no acquisition of KP772 resistance. Summarizing, our data demonstrate that KP772 is hyperactive in MDR cells and might have chemosensitizing properties by blocking ABCB1 expression. Together with the disability of tumor cells to acquire KP772 resistance, our data suggest that KP772 should be especially active against notoriously drug-resistant tumor types and as second line treatment after standard chemotherapy failure.


Assuntos
Antineoplásicos/farmacologia , Resistência a Múltiplos Medicamentos , Lantânio/farmacologia , Compostos Organometálicos/farmacologia , Fenantrolinas/farmacologia , Subfamília B de Transportador de Cassetes de Ligação de ATP , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Adenocarcinoma/tratamento farmacológico , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Carcinoma de Células Pequenas/tratamento farmacológico , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos , Formazans/metabolismo , Células HL-60 , Humanos , Lantânio/química , Lantânio/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Estrutura Molecular , Proteínas de Neoplasias/metabolismo , Transportadores de Ânions Orgânicos/metabolismo , Compostos Organometálicos/química , Compostos Organometálicos/uso terapêutico , Fenantrolinas/química , Fenantrolinas/uso terapêutico , Sensibilidade e Especificidade , Sais de Tetrazólio/metabolismo
19.
Rev Physiol Biochem Pharmacol ; 153: 101-11, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15674649

RESUMO

Cerium is a member of the lanthanide series or rare earth elements which exert diverse biological effects mainly by their resemblance to calcium. This similarity, which is particularly characteristic for the lighter members of the lanthanide series, enables these elements to replace calcium in biomolecules without necessarily substituting for it functionally. While the inhibitory effects on calcium-dependent physiological processes (such as those involved in the blood clotting cascade as well as in neuronal and muscular functions) are well-known, their relevance for the pharmacological properties of cerium are less clear. Historically, cerium oxalate was used as an antiemetic, especially in vomiting of pregnancy and kinetoses, although its mechanism of action has never been clarified. At present, cerium nitrate is available as an adjunct to silver sulfadiazine cream for the topical treatment of extensive burns not amenable to early wound excision. Apart from direct antiseptic effects, cerium helps to prevent postburn sepsis and systemic inflammatory response by fixing burn toxins. The antineoplastic potential of cerium compounds, which had fallen into oblivion, is currently being re-explored in experimental settings, though the mechanistic basis remains to be elucidated.


Assuntos
Queimaduras/tratamento farmacológico , Cério , Neoplasias/tratamento farmacológico , Vômito/tratamento farmacológico , Animais , Disponibilidade Biológica , Queimaduras/metabolismo , Cério/farmacocinética , Cério/farmacologia , Cério/uso terapêutico , Humanos , Neoplasias/metabolismo , Vômito/metabolismo
20.
J Pharmacol Exp Ther ; 312(1): 281-9, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15331656

RESUMO

KP1019 [indazolium trans-[tetrachlorobis(1H-indazole)ruthenate (III)] (FFC14A) is a metal complex with promising anticancer activity. Since chemoresistance is a major obstacle in chemotherapy, this study investigated the influence of several drug resistance mechanisms on the anticancer activity of KP1019. Here we demonstrate that the cytotoxic effects of KP1019 are neither substantially hampered by overexpression of the drug resistance proteins multidrug resistance-related protein 1, breast cancer resistance protein, and lung resistance protein nor the transferrin receptor and only marginally by the cellular p53 status. In contrast, P-glycoprotein overexpression weakly but significantly (up to 2-fold) reduced KP1019 activity. P-glycoprotein-related resistance was based on reduced intracellular KP1019 accumulation and reversible by known P-glycoprotein modulators. KP1019 dose dependently inhibited ATPase activity of P-glycoprotein with a K(i) of approximately 31 microM. Furthermore, it potently blocked P-glycoprotein-mediated rhodamine 123 efflux under serum-free conditions (EC(50), approximately 8 microM), however, with reduced activity at increased serum concentrations (EC(50) at 10% serum, approximately 35 microM). Moreover, P-glycoprotein-mediated daunomycin resistance could only be marginally restored by KP1019 in serum-containing medium, also indicating an influence of serum proteins on the interaction between KP1019 and P-glycoprotein. Acquired KP1019 resistance was investigated by selecting KB-3-1 cells against KP1019 for more than 1 year. Only an approximately 2-fold KP1019 resistance could be induced, which unexpectedly was not due to overexpression of P-glycoprotein or other efflux pumps. Accordingly, KP1019-resistant cells did not display reduced drug accumulation. Their unique cross-resistance pattern confirmed an ABC transporter-independent resistance phenotype. In summary, the likeliness of acquiring insensitivity to KP1019 during therapy is expected to be low, and resistance should not be based on overexpression of drug efflux transporters.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Indazóis/farmacologia , Compostos de Rutênio/farmacologia , Adenosina Trifosfatases/metabolismo , Genes MDR/fisiologia , Células HL-60 , Humanos , Células KB , Compostos Organometálicos , Receptores da Transferrina/metabolismo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA