Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Inorg Chem ; 60(23): 18048-18057, 2021 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-34781677

RESUMO

Gly-His-Lys (GHK) is a tripeptide present in the human bloodstream that exhibits a number of biological functions. Its activity is attributed to the copper-complexed form, Cu(II)GHK. Little is known, however, about the molecular aspects of the mechanism of its action. Here, we examined the reaction of Cu(II)GHK with reduced glutathione (GSH), which is the strongest reductant naturally occurring in human plasma. Spectroscopic techniques (UV-vis, CD, EPR, and NMR) and cyclic voltammetry helped unravel the reaction mechanism. The impact of temperature, GSH concentration, oxygen access, and the presence of ternary ligands on the reaction were explored. The transient GSH-Cu(II)GHK complex was found to be an important reaction intermediate. The kinetic and redox properties of this complex, including tuning of the reduction rate by ternary ligands, suggest that it may provide a missing link in copper trafficking as a precursor of Cu(I) ions, for example, for their acquisition by the CTR1 cellular copper transporter.


Assuntos
Complexos de Coordenação/metabolismo , Cobre/metabolismo , Glutationa/metabolismo , Oligopeptídeos/metabolismo , Compostos de Sulfidrila/metabolismo , Complexos de Coordenação/sangue , Complexos de Coordenação/química , Cobre/sangue , Cobre/química , Glutationa/sangue , Glutationa/química , Humanos , Estrutura Molecular , Oligopeptídeos/sangue , Oligopeptídeos/química , Oxirredução , Compostos de Sulfidrila/sangue , Compostos de Sulfidrila/química
2.
Curr Top Med Chem ; 21(1): 48-58, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32600232

RESUMO

Although metallodrugs are used to treat a variety of human disorders and exhibit a remarkable diversity of therapeutic properties, they constitute only a tiny minority of all medicinal drugs that are currently on the market. This undesirable situation must be partially attributed to our general lack of understanding the fate of metallodrugs in the extremely ligand-rich environment of the bloodstream. The challenge of gaining insight into these bioinorganic processes can be overcome by the application of 'metallomics tools', which involve the analysis of biological fluids (e.g., blood plasma) with a separation method in conjunction with multi-element specific detectors. To this end, we have developed a metallomics tool that is based on size-exclusion chromatography (SEC) hyphenated to an inductively coupled plasma atomic emission spectrometer (ICP-AES). After the successful application of SEC-ICPAES to analyze plasma for endogenous copper, iron and zinc-metalloproteins, it was subsequently applied to probe the metabolism of a variety of metal-based anticancer drugs in plasma. The versatility of this metallomics tool is exemplified by the fact that it has provided insight into the metabolism of individual Pt-based drugs, the modulation of the metabolism of cisplatin by sulfur-containing compounds, the metabolism of two metal-based drugs that contain different metals as well as a bimetallic anticancer drug, which contained two different metals. After adding pharmacologically relevant doses of metallodrugs to plasma, the temporal analysis of aliquots by SEC-ICP-AES allows to observe metal-protein adducts, metallodrug-derived degradation products and the parent metallodrug(s). This unique capability allows to obtain comprehensive insight into the fate of metal-based drugs in plasma and can be extended to in vivo studies. Thus, the application of this metallomics tool to probe the fate of novel metalcomplexes that exert the desired biological activity in plasma has the potential to advance more of these to animal/preclinical studies to fully explore the potential that metallodrugs inherently offer.


Assuntos
Antineoplásicos/farmacologia , Complexos de Coordenação/farmacologia , Metaloproteínas/metabolismo , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/sangue , Antineoplásicos/química , Proliferação de Células/efeitos dos fármacos , Complexos de Coordenação/sangue , Complexos de Coordenação/química , Humanos , Metaloproteínas/sangue , Metaloproteínas/química , Neoplasias/sangue , Neoplasias/patologia
3.
Molecules ; 24(18)2019 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-31514445

RESUMO

The current study aimed to synthesize new metal coordination complexes with potential biomedical applications. Metal complexes were prepared via the reaction of isatin-N(4)anti- pyrinethiosemicarbazone ligand 1 with Cu(II), Ni(II), Co(II), Zn(II), and Fe(III) ions. The obtained metal complexes 2-12 were characterized using elemental, spectral (1H-NMR, EPR, Mass, IR, UV-Vis) and thermal (TGA) techniques, as well as magnetic moment and molar conductance measurements. In addition, their geometries were studied using EPR and UV-Vis spectroscopy. To evaluate the in vivo anti-cancer activities of these complexes, the ligand 1 and its metal complexes 2, 7 and 9 were tested against solid tumors. The solid tumors were induced by subcutaneous (SC) injection of Ehrlich ascites carcinoma (EAC) cells in mice. The impact of the selected complexes on the reduction of tumor volume was determined. Also, the expression levels of vascular endothelial growth factor (VEGF) and cysteine aspartyl-specific protease-7 (caspase-7) in tumor and liver tissues of mice bearing EAC tumor were determined. Moreover, their effects on alanine transaminase (ALT), aspartate transaminase (AST), albumin, and glucose levels were measured. The results revealed that the tested compounds, especially complex 9, reduced tumor volume, inhibited the expression of VEGF, and induced the expression of caspase-7. Additionally, they restored the levels of ALT, AST, albumin, and glucose close to their normal levels. Taken together, our newly synthesized metal complexes are promising anti-cancer agents against solid tumors induced by EAC cells as supported by the inhibition of VEGF and induction of caspase-7.


Assuntos
Antineoplásicos/síntese química , Antineoplásicos/uso terapêutico , Ascite/tratamento farmacológico , Carcinoma de Ehrlich/tratamento farmacológico , Complexos de Coordenação/síntese química , Complexos de Coordenação/uso terapêutico , Isatina/uso terapêutico , Animais , Antineoplásicos/farmacologia , Ascite/sangue , Carcinoma de Ehrlich/sangue , Complexos de Coordenação/sangue , Complexos de Coordenação/farmacologia , Espectroscopia de Ressonância de Spin Eletrônica , Elétrons , Feminino , Isatina/química , Isatina/farmacologia , Ligantes , Camundongos , Espectroscopia de Prótons por Ressonância Magnética , Espectrofotometria Infravermelho , Temperatura
4.
Anal Chem ; 91(5): 3681-3686, 2019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30698003

RESUMO

A sensitive electrochemiluminescence (ECL) biosensor was developed for glutathione (GSH) detection based on a novel Ru(bpy)2(cpaphen)2+/TPrA/TiO2 ternary ECL system with Mn2+ as substitute target for signal amplification. Specifically, the TiO2 nanoneedles (TiO2 NNs) were used as the coreaction accelerator for the first time to promote the oxidation process of coreactant tripropylamine (TPrA) in the anode and significantly increase the ECL signal of Ru(bpy)2(cpaphen)2+ for an amplified initial signal. Meanwhile, a novel target conversion strategy for GSH was developed by reducing MnO2 nanosheets to Mn2+ as a substitute target, which played the role of a coenzyme factor for cleaving DNA double strands intercalated with Ru(bpy)2(cpaphen)2+ to markedly weaken initial signal. As a result, the novel "on-off" biosensor achieved a sensitive detection of GSH range from 5 µM to 215 µM with a detection limit of 0.33 µM. Importantly, the proposed strategy enriched the application of Ru complex and TPrA ECL system in bioanalytical applications, and provided a new signal amplification strategy for bioactive small molecules.


Assuntos
Técnicas Biossensoriais/métodos , Complexos de Coordenação/sangue , Glutationa/análise , Poliaminas/química , Rutênio/química , Titânio/química , Técnicas Biossensoriais/normas , Quebras de DNA de Cadeia Dupla , Técnicas Eletroquímicas , Substâncias Intercalantes , Limite de Detecção , Manganês
5.
Proc Natl Acad Sci U S A ; 115(45): 11454-11459, 2018 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-30348798

RESUMO

Bioimaging plays an important role in cancer diagnosis and treatment. However, imaging sensitivity and specificity still constitute key challenges. Nanotechnology-based imaging is particularly promising for overcoming these limitations because nanosized imaging agents can specifically home in on tumors via the "enhanced permeation and retention" (EPR) effect, thus resulting in enhanced imaging sensitivity and specificity. Here, we report an original nanosystem for positron emission tomography (PET) imaging based on an amphiphilic dendrimer, which bears multiple PET reporting units at the terminals. This dendrimer is able to self-assemble into small and uniform nanomicelles, which accumulate in tumors for effective PET imaging. Benefiting from the combined dendrimeric multivalence and EPR-mediated passive tumor targeting, this nanosystem demonstrates superior imaging sensitivity and specificity, with up to 14-fold increased PET signal ratios compared with the clinical gold reference 2-fluorodeoxyglucose ([18F]FDG). Most importantly, this dendrimer system can detect imaging-refractory low-glucose-uptake tumors that are otherwise undetectable using [18F]FDG. In addition, it is endowed with an excellent safety profile and favorable pharmacokinetics for PET imaging. Consequently, this dendrimer nanosystem constitutes an effective and promising approach for cancer imaging. Our study also demonstrates that nanotechnology based on self-assembling dendrimers provides a fresh perspective for biomedical imaging and cancer diagnosis.


Assuntos
Neoplasias do Colo/diagnóstico por imagem , Complexos de Coordenação/farmacocinética , Radioisótopos de Gálio/farmacocinética , Glioblastoma/diagnóstico por imagem , Neoplasias Pancreáticas/diagnóstico por imagem , Tomografia por Emissão de Pósitrons/métodos , Neoplasias da Próstata/diagnóstico por imagem , Animais , Linhagem Celular Tumoral , Neoplasias do Colo/patologia , Meios de Contraste/química , Meios de Contraste/farmacocinética , Complexos de Coordenação/sangue , Complexos de Coordenação/química , Dendrímeros/química , Fluordesoxiglucose F18/química , Radioisótopos de Gálio/sangue , Radioisótopos de Gálio/química , Glioblastoma/patologia , Compostos Heterocíclicos/química , Compostos Heterocíclicos com 1 Anel , Xenoenxertos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Pancreáticas/patologia , Neoplasias da Próstata/patologia
6.
Dalton Trans ; 47(15): 5252-5258, 2018 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-29560976

RESUMO

The potential advantage of platinum(iv) complexes as alternatives to classical platinum(ii)-based drugs relies on their kinetic stability in the body before reaching the tumor site and on their activation by reduction inside cancer cells. In this study, an analytical workflow has been developed to investigate the reductive biotransformation and kinetic inertness of platinum(iv) prodrugs comprising different ligand coordination spheres (respectively, lipophilicity and redox behavior) in whole human blood. The distribution of platinum(iv) complexes in blood pellets and plasma was determined by inductively coupled plasma-mass spectrometry (ICP-MS) after microwave digestion. An analytical approach based on reversed-phase (RP)-ICP-MS was used to monitor the parent compound and the formation of metabolites using two different extraction procedures. The ligand coordination sphere of the platinum(iv) complexes had a significant impact on their accumulation in red blood cells and on their degree of kinetic inertness in whole human blood. The most lipophilic platinum(iv) compound featuring equatorial chlorido ligands showed a pronounced penetration into blood cells and a rapid reductive biotransformation. In contrast, the more hydrophilic platinum(iv) complexes with a carboplatin- and oxaliplatin-core exerted kinetic inertness on a pharmacologically relevant time scale with notable amounts of the compound accumulated in the plasma fraction.


Assuntos
Carboplatina/sangue , Carboplatina/farmacocinética , Complexos de Coordenação/sangue , Complexos de Coordenação/farmacocinética , Compostos Organoplatínicos/sangue , Compostos Organoplatínicos/farmacocinética , Pró-Fármacos/farmacocinética , Cromatografia Líquida de Alta Pressão , Humanos , Interações Hidrofóbicas e Hidrofílicas , Cinética , Ligantes , Nanosferas/química , Oxaliplatina , Oxirredução
7.
J Inorg Biochem ; 181: 87-95, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29307441

RESUMO

We reviewed the three toxicokinetic reference studies commonly used to suggest that aluminum (Al)-based adjuvants are innocuous. A single experimental study was carried out using isotopic 26Al (Flarend et al., Vaccine, 1997). This study used aluminum salts resembling those used in vaccines but ignored adjuvant uptake by cells that was not fully documented at the time. It was conducted over a short period of time (28days) and used only two rabbits per adjuvant. At the endpoint, Al elimination in the urine accounted for 6% for Al hydroxide and 22% for Al phosphate, both results being incompatible with rapid elimination of vaccine-derived Al in urine. Two theoretical studies have evaluated the potential risk of vaccine Al in infants, by reference to an oral "minimal risk level" (MRL) extrapolated from animal studies. Keith et al. (Vaccine, 2002) used a high MRL (2mg/kg/d), an erroneous model of 100% immediate absorption of vaccine Al, and did not consider renal and blood-brain barrier immaturity. Mitkus et al. (Vaccine, 2011) only considered solubilized Al, with erroneous calculations of absorption duration. Systemic Al particle diffusion and neuro-inflammatory potential were omitted. The MRL they used was both inappropriate (oral Al vs. injected adjuvant) and still too high (1mg/kg/d) regarding recent animal studies. Both paucity and serious weaknesses of reference studies strongly suggest that novel experimental studies of Al adjuvants toxicokinetics should be performed on the long-term, including both neonatal and adult exposures, to ensure their safety and restore population confidence in Al-containing vaccines.


Assuntos
Adjuvantes Imunológicos/efeitos adversos , Compostos de Alumínio/efeitos adversos , Alumínio/toxicidade , Complexos de Coordenação/toxicidade , Vacinas/efeitos adversos , Absorção Fisiológica , Adjuvantes Imunológicos/sangue , Adjuvantes Imunológicos/metabolismo , Adjuvantes Imunológicos/farmacocinética , Adolescente , Adulto , Fatores Etários , Alumínio/sangue , Alumínio/metabolismo , Alumínio/urina , Compostos de Alumínio/sangue , Compostos de Alumínio/metabolismo , Compostos de Alumínio/farmacocinética , Animais , Criança , Complexos de Coordenação/sangue , Complexos de Coordenação/metabolismo , Complexos de Coordenação/urina , Humanos , Lactente , Eliminação Renal , Testes de Toxicidade , Toxicocinética
8.
Anticancer Drugs ; 28(8): 898-910, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28657910

RESUMO

The main aim of this study is to assess the safety and antitumor efficacy of a palladium(II) (Pd)-saccharinate complex with terpyridine. To characterize the Pd(II) complex in vitro, its cytotoxicity was evaluated using a water-soluble tetrazolium salt cell viability assay and the mechanism of cell death was assessed by DNA fragmentation/condensation and live cell imaging analyses. The antitumor efficacy and safety of the Pd(II) complex in-vivo were examined by analyzing reduction in tumor size, changes in body and organ weight, histopathological analysis of liver, kidney, and tumor sections, and biochemical analysis of serum in C57BL/6 mice. Our results showed that the Pd(II) complex was more cytotoxic to cancer cells than noncancer cell lines and caused cell death through apoptotic pathways. The treatment of the Pd(II) complex in tumor-bearing mice effectively reduced the tumor size at half the dose used for cisplatin. The Pd(II) complex appeared to exert less liver damage than the cisplatin-based complex on changes in the hepatic enzymes levels in the serum. Hence, the complex appears to be a potential chemotherapeutic drug with high antitumor efficacy and fewer hepatotoxic complications, providing an avenue for further studies.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Cisplatino/farmacologia , Complexos de Coordenação/química , Complexos de Coordenação/farmacologia , Neoplasias/tratamento farmacológico , Células A549 , Aloenxertos , Animais , Antineoplásicos/sangue , Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Cisplatino/sangue , Cisplatino/toxicidade , Complexos de Coordenação/sangue , Complexos de Coordenação/toxicidade , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Células HeLa , Humanos , Fígado/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Neoplasias/sangue
9.
Chemistry ; 23(41): 9888-9896, 2017 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-28509422

RESUMO

Two [Ru(phen)2 dppz]2+ derivatives (phen=1,10-phenantroline, dppz=dipyrido[3,2-a:2',3'-c]phenazine) with different functional groups on the dppz ligand [dppz-7,8-(OMe)2 (1), dppz-7,8-(OH)2 (2)] have been synthesized, characterized and investigated as photosensitizers (PSs) for photodynamic therapy (PDT) against cancer. Both complexes showed intense red phosphorescence and promising singlet oxygen (1 O2 ) quantum yields of 75 % (1) and 54 % (2) in acetonitrile. Complex 1 (logPo/w =-0.52, 2.4 nmol Ru per mg protein) was found to be more lipophilic, having also a higher cellular uptake efficiency compared to 2 (logPo/w =-0.20, 0.9 nmol Ru per mg protein). Complex 1 localized evenly in HeLa cells whereas 2, was mainly visualized in the cell membrane by confocal microscopy. In the dark, complex 1 (IC50 =36.5 µm) was found to be more toxic than complex 2 (IC50 >100 µm) on a HeLa cells monolayer. Importantly, in view of PDT applications, both complexes were found to be non-toxic in the dark towards multicellular HeLa spheroids (IC50 >100 µm). Upon one-photon irradiation (420 nm, 9.27 J cm-2 ), 1 exhibited higher phototoxicity (IC50 =3.1 µm) than 2 (IC50 =16.7 µm) on HeLa cell monolayers. When two-photon irradiation (800 nm, 9.90 J cm-2 ) was applied, only 1 (IC50 =9.5 µm) was found to be active toward HeLa spheroids. This study demonstrates that the functional group on the intercalative ligand has a strong influence on the cellular localization and anticancer activity of RuII polypyridyl complexes.


Assuntos
Complexos de Coordenação/química , Fármacos Fotossensibilizantes/química , Rutênio/química , Sobrevivência Celular/efeitos dos fármacos , Complexos de Coordenação/sangue , Complexos de Coordenação/síntese química , Complexos de Coordenação/toxicidade , Cristalografia por Raios X , Células HeLa , Humanos , Interações Hidrofóbicas e Hidrofílicas , Luz , Microscopia Confocal , Conformação Molecular , Compostos Organometálicos/química , Fotoquimioterapia , Fótons , Fármacos Fotossensibilizantes/sangue , Fármacos Fotossensibilizantes/síntese química , Fármacos Fotossensibilizantes/toxicidade , Oxigênio Singlete/química , Oxigênio Singlete/metabolismo , Esferoides Celulares/citologia , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/metabolismo
10.
Chembiochem ; 18(12): 1083-1086, 2017 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-28425643

RESUMO

Chemical (as opposed to light-induced) activation of caged molecules is a rapidly advancing approach to trigger biological processes. We previously introduced the ruthenium-catalyzed release of allyloxycarbonyl (alloc)-protected amines in human cells. A restriction of this and all other methods is the limited lifetime of the catalyst, thus hampering meaningful applications. In this study, we addressed this problem with the development of a new generation of ruthenium complexes for the uncaging of alloc-protected amines with superior catalytic activity. Under biologically relevant conditions, we achieved a turnover number >300, a reaction rate of 580 m-1 s-1 , and we observed high activity in blood serum. Furthermore, alloc-protected doxorubicin, as an anticancer prodrug, could be activated in human cell culture and induced apoptosis with a single low dose (1 µm) of the new catalyst.


Assuntos
Compostos Alílicos/química , Aminas/química , Complexos de Coordenação/síntese química , Doxorrubicina/agonistas , Rutênio/química , Antibióticos Antineoplásicos/sangue , Antibióticos Antineoplásicos/síntese química , Antibióticos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Catálise , Complexos de Coordenação/sangue , Complexos de Coordenação/química , Doxorrubicina/análogos & derivados , Doxorrubicina/sangue , Doxorrubicina/farmacologia , Células HeLa , Humanos , Concentração Inibidora 50 , Pró-Fármacos/síntese química , Pró-Fármacos/farmacologia , Rutênio/sangue
12.
J Inorg Biochem ; 160: 61-9, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26507768

RESUMO

Triapine has been investigated as anticancer drug in multiple clinical phase I/II trials. Although promising anti-leukemic activity was observed, Triapine was ineffective against solid tumors. The reasons are currently widely unknown. The biological activity of Triapine is strongly connected to its iron complex (Fe-Triapine) which is pharmacologically not investigated. Here, novel analytical tools for Triapine and Fe-Triapine were developed and applied for cell extracts and body fluids of treated mice. Triapine and its iron complex showed a completely different behavior: for Triapine, low protein binding was observed in contrast to fast protein adduct formation of Fe-Triapine. Notably, both drugs were rapidly cleared from the body (serum half-life time <1h). Remarkably, in contrast to Triapine, where (in accordance to clinical data) basically no renal excretion was found, the iron complex was effectively excreted via urine. Moreover, no Fe-Triapine was detected in serum or cytosolic extracts after Triapine treatment. Taken together, our study will help to further understand the biological behavior of Triapine and its Fe-complex and allow the development of novel thiosemicarbazones with pronounced activity against solid tumor types.


Assuntos
Antineoplásicos/farmacocinética , Neoplasias do Colo/tratamento farmacológico , Complexos de Coordenação/farmacocinética , Ferro/farmacocinética , Piridinas/farmacocinética , Tiossemicarbazonas/farmacocinética , Animais , Antineoplásicos/sangue , Antineoplásicos/urina , Proteínas Sanguíneas/química , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ensaios Clínicos como Assunto , Neoplasias do Colo/patologia , Complexos de Coordenação/sangue , Complexos de Coordenação/urina , Feminino , Meia-Vida , Ferro/sangue , Ferro/urina , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Ligação Proteica , Piridinas/sangue , Piridinas/urina , Tiossemicarbazonas/sangue , Tiossemicarbazonas/urina , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Biomaterials ; 75: 47-57, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26480471

RESUMO

A novel MRI blood-pool contrast agent (Gd-AAZTA-MADEC) has been compared with established blood pool agents for tumor contrast enhanced images and angiography. Synthesis, relaxometric properties, albumin binding affinity and pharmacokinetic profiles are reported. For in vivo studies, angiographic images and tumor contrast enhanced images were acquired on mice with benchtop 1T-MRI scanners and compared with MS-325, B22956/1 and B25716/1. The design of this contrast agent involved the elongation of the spacer between the targeting deoxycholic acid moiety and the Gd-AAZTA imaging reporting unit that drastically changed either the binding affinity to albumin (KA(HSA) = 8.3 × 10(5) M(-1)) and the hydration state of the Gd ion (q = 2) in comparison to the recently reported B25716/1. The very markedly high binding affinity towards mouse and human serum albumins resulted in peculiar pharmacokinetics and relaxometric properties. The NMRD profiles clearly indicated that maximum efficiency is attainable at magnetic field strength of 1 T. In vivo studies showed high enhancement of the vasculature and a prolonged accumulation inside tumor. The herein reported pre-clinical imaging studies show that a great benefit arises from the combination of a benchtop MRI scanner operating at 1 T and the albumin-binding Gd-AAZTA-MADEC complex, for pursuing enhanced angiography and improved characterization of tumor vascular microenvironment.


Assuntos
Ácidos Cólicos/química , Meios de Contraste/química , Complexos de Coordenação/química , Imagem do Acúmulo Cardíaco de Comporta , Imageamento por Ressonância Magnética/instrumentação , Animais , Ácidos Cólicos/sangue , Ácidos Cólicos/síntese química , Ácidos Cólicos/farmacocinética , Complexos de Coordenação/sangue , Complexos de Coordenação/síntese química , Complexos de Coordenação/farmacocinética , Humanos , Angiografia por Ressonância Magnética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neoplasias/diagnóstico , Ligação Proteica , Espectroscopia de Prótons por Ressonância Magnética , Prótons , Albumina Sérica/metabolismo , Distribuição Tecidual , Água
14.
Drug Discov Today Technol ; 16: 24-30, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26547418

RESUMO

Understanding the fate of metallodrugs in the bloodstream is critical to assess if the parent drug has a reasonable probability to reach the intended target tissue and to predict toxic side-effects. To gain insight into these processes, we have added pharmacologically relevant doses of metallodrugs to blood plasma and applied an LC-method to directly analyze the latter for metallodrug metabolites. Using human or rabbit plasma, this LC-method was employed to gain insight into the metabolism of clinically used as well as emerging anticancer metallodrugs and to unravel the mechanisms by which small molecular weight compounds that - when co-administered with a metallodrug - decrease the toxic side-effects of the metallodrug by modulating its metabolism. The results suggest that the developed LC-method is useful to probe the fate of biologically active novel metal-complexes in plasma to help select those which may be advanced to animal/clinical studies to ultimately develop safer metallodrugs.


Assuntos
Antineoplásicos/sangue , Antineoplásicos/metabolismo , Complexos de Coordenação/sangue , Complexos de Coordenação/metabolismo , Animais , Cromatografia Líquida/métodos , Humanos , Ligação Proteica/fisiologia , Coelhos
15.
J Inorg Biochem ; 153: 167-177, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26281973

RESUMO

The biotransformation in the plasma and red blood cells of two potential antitumor V(IV)O complexes formed by flavonoid ligands (quercetin or que and morin or mor) and their sulfonic derivatives (quercetin-5'-sulfonic acid or que(S) and morin-5'-sulfonic acid or mor(S)) was studied by spectroscopic (EPR, Electron Paramagnetic Resonance) and computational (DFT, Density Functional Theory) methods. Que and que(S) form with V(IV)O stable complexes, and in the systems with apo-transferrin (apo-hTf) and albumin (HSA) VO(que)2 and VO(que(S))2 remain unchanged. VO(mor)2 and VO(mor(S))2 undergo displacement reactions to give the partial formation of (VO)x(HSA) and (VO)(apo-hTf)/(VO)2(apo-hTf); moreover, mor(S) forms with apo-transferrin and albumin mixed species VO-mor(S)-apo-hTf and VO-mor(S)-HSA. In the systems with apo-hTf and HSA anisotropic EPR spectra at room temperature are detected in which the protein is not directly coordinated to V(IV)O(2+) ion. This is explained assuming that the bis-chelated complexes interact strongly with the proteins through a network of hydrogen bonds with the polar groups present on the protein surface. It is suggested that this "indirect" transport of V(IV)O species could be common to all the species containing ligands which can interact with the blood proteins. Uptake experiments by red blood cells were also carried out, using vanadium concentration of 5.0×10(-4)M and incubation time in the range 0-160min. VO(que)2/VO(que(S))2 and VO(mor)2/VO(mor(S))2 cross the erythrocytes membrane and in the cytosol VO(que)2/VO(que(S))2 do not transform, whereas VO(mor)2/VO(mor(S))2 give the partial formation of mixed species with hemoglobin (Hb) and other V(IV)O complexes.


Assuntos
Complexos de Coordenação/química , Flavonoides/química , Quercetina/química , Ácidos Sulfônicos/química , Vanádio/química , Apoproteínas/química , Biotransformação , Ácido Cítrico/química , Complexos de Coordenação/sangue , Complexos de Coordenação/síntese química , Eritrócitos/metabolismo , Flavonoides/sangue , Flavonoides/síntese química , Hemoglobinas/química , Humanos , Ligação de Hidrogênio , Ácido Láctico/química , Ligantes , Modelos Químicos , Quercetina/sangue , Quercetina/síntese química , Albumina Sérica/química , Ácidos Sulfônicos/sangue , Ácidos Sulfônicos/síntese química , Transferrina/química
16.
Bioorg Med Chem Lett ; 25(5): 1082-5, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25661712

RESUMO

A promising bifunctional chelate (N-NE3TA) was conjugated to bile acids, cholic acid (CA), deoxycholic acid (DCA), and chenodeoxycholic acid (CDCA) as tumor targeting vectors. Bile acid conjugates of N-NE3TA (CA-N-NE3TA, DCA-N-NE3TA, and CDCA-N-NE3TA) were comparatively evaluated for complexation with (64)Cu, an imaging probe for positron emission tomography (PET). N-NE3TA-bile acid conjugates were evaluated for radiolabeling kinetics with (64)Cu, and the corresponding (64)Cu-radiolabeled conjugates were screened for complex stability in human serum and EDTA solution. The NE3TA-bile acid conjugates instantly bound to (64)Cu with excellent radiolabeling efficiency at room temperature. All NE3TA-bile acid conjugates radiolabeled with (64)Cu remained inert in human serum for 2days without releasing a considerable amount of the radioactivity. The (64)Cu-radiolabeled complexes were further challenged by EDTA in a 100-fold molar excess. Bile acid-N-NE3TA conjugates radiolabeled with (64)Cu were quite stable with a minimal transfer of (64)Cu to EDTA at 4h time point. The in vitro data indicate that the bile acid-N-NE3TA conjugates deserve further biological evaluation for (64)Cu-based targeted PET imaging applications.


Assuntos
Ácidos e Sais Biliares/química , Complexos de Coordenação/química , Radioisótopos de Cobre/química , Tomografia por Emissão de Pósitrons/métodos , Ácidos e Sais Biliares/sangue , Complexos de Coordenação/sangue , Radioisótopos de Cobre/sangue , Humanos , Compostos Radiofarmacêuticos/sangue , Compostos Radiofarmacêuticos/química
17.
Molecules ; 19(8): 12194-223, 2014 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-25123186

RESUMO

The speciation of Cu2+, Ni2+ and Zn2+ ions in the presence of the fluoroquinolones (FQs) moxifloxacin, ofloxacin, levofloxacin and ciprofloxacin, in human blood plasma was studied under physiological conditions by computer simulation. The speciation was calculated using an updated model of human blood plasma including over 6,000 species with the aid of the program Hyss2009. The identity and stability of metal-FQ complexes were determined by potentiometric (310 K, 0.15 mol/L NaCl), spectrophotometric, spectrofluorimetric, ESI-MS and 1H-NMR measurements. In the case of Cu2+ ion the concentration of main low molecular weight (LMW) plasma complex (Cu(Cis)His) is very slightly influenced by all examined FQs. FQs show much higher influence on main plasma Ni2+ and Zn2+ complexes: (Ni(His)2 and Zn(Cys)Cit, respectively. Levofloxacin exhibits the highest influence on the fraction of the main nickel complex, Ni(His)2, even at a concentration level of 3×10⁻5 mol/L. The same effect is seen on the main zinc complex, Zn(Cys)Cit. Calculated plasma mobilizing indexes indicate that ciprofloxacin possesses the highest mobilizing power from plasma proteins, toward copper ion, while levofloxacin is the most influential on nickel and zinc ions. The results obtained indicate that the drugs studied are safe in relation to mobilization of essential metal ions under physiological conditions. The observed effects were explained in terms of competitive equilibrium reactions between the FQs and the main LMW complexes of the metal ions.


Assuntos
Cobre/sangue , Fluoroquinolonas/sangue , Íons/sangue , Níquel/sangue , Zinco/sangue , Ciprofloxacina/química , Ciprofloxacina/isolamento & purificação , Complexos de Coordenação/sangue , Complexos de Coordenação/química , Cobre/química , Fluoroquinolonas/química , Humanos , Levofloxacino/sangue , Levofloxacino/química , Moxifloxacina , Níquel/química , Ofloxacino/sangue , Ofloxacino/química , Zinco/química
18.
Talanta ; 117: 412-8, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-24209361

RESUMO

New lanthanide complex Gd-ANAMD containing 2-amino-7-methyl-1,8-naphthyridine was achieved for selective magnetic resonance imaging towards guanosine 5'-monophosphate over other ribonucleotide polyphosphates in aqueous media and in vivo. The formation of strong multi-hydrogen bonds between naphthyridine and guanosine made the phosphate in guanosine 5'-monophosphate positioned on a suitable site to coordinate with the lanthanide ion. The substitution of the coordination naphthyridine by the phosphate oxygen atoms caused obvious relaxivity decrease. The negligible cytotoxicity and appropriate blood circulation time of Gd-ANAMD allow potential application of Magnetic Resonance Imaging in vivo. (1)H NMR confirmed that the selectivity of these lanthanide complexes towards guanosine was attributed to the formation of hydrogen bonds between the guanine moeity and the naphthyridine. The fluorescence detection and lifetime measurement of Tb-ANAMD and Eu-ANAMD suggested that the decrease of the relaxivity is not attributed to the change of the q value, but caused by the prolonging of the residence lifetime of inner-sphere water.


Assuntos
Meios de Contraste/química , Complexos de Coordenação/química , Gadolínio/química , Guanosina Monofosfato/análise , Imageamento por Ressonância Magnética/métodos , Naftiridinas/química , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Meios de Contraste/metabolismo , Meios de Contraste/farmacocinética , Complexos de Coordenação/sangue , Complexos de Coordenação/farmacocinética , Cristalografia por Raios X , Humanos , Ligação de Hidrogênio , Injeções Intraperitoneais , Injeções Subcutâneas , Espectroscopia de Ressonância Magnética , Camundongos , Termodinâmica , Água/química
19.
Nucl Med Biol ; 40(1): 33-41, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22995902

RESUMO

Gallium-68 is rapidly gaining importance, as this generator-produced PET isotope is available independent of on-site cyclotrons, enabling radiopharmaceutical production with comparably simple techniques at low cost. The recently introduced TRAP chelator combines the advantage of straightforward design of multimeric 68Ga-radiopharmaceuticals with very fast and efficient 68Ga-labeling. We synthesized a series of five cyclo(RGDfK) peptide trimers and determined their α(v)ß3 integrin affinities in competition assays on α(v)ß3-expressing M21 human melanoma cells against ¹²5I-echistatin. The compound with highest IC50, Ga-TRAP(RGD)3, showed more than 7-fold higher affinity compared to the monomers F-Galacto-RGD and Ga-NODAGA-c(RGDyK). TRAP(RGD)3 was radiolabeled with 68Ga in a fully automated GMP compliant manner. CD-1 athymic nude mice bearing M21/M21L human melanoma xenografts were used for biodistribution studies, blockade experiments, metabolite studies and PET imaging. 68Ga-TRAP(RGD)3 exhibited high M21 tumor uptake (6.08±0.63% ID/g, 60 min p.i.), was found to be fully stable in vivo, and showed a fast renal clearance. Blockade studies showed that uptake in the tumor, as well as in all other tissues, is highly integrin specific. A comparison of biodistribution and PET data of 68Ga-TRAP(RGD)3 with those of 68Ga-NODAGA-c(RGDyK) and ¹8F-Galacto-RGD showed that the higher affinity of the trimer effects a larger dynamic response of tracer uptake to integrin expression, i.e., enhanced integrin-specific uptake in all tissues. We conclude that 68Ga-TRAP(RGD)3 could allow for imaging of low-level integrin expression in tissues which are not visible with the two competitors. Overall, the study constitutes proof of concept for the favourable in vivo properties of TRAP-based 68Ga radiopharmaceuticals.


Assuntos
Complexos de Coordenação/química , Oligopeptídeos/química , Compostos Organofosforados/química , Peptídeos Cíclicos/química , Animais , Complexos de Coordenação/sangue , Complexos de Coordenação/farmacocinética , Estabilidade de Medicamentos , Feminino , Radioisótopos de Gálio , Humanos , Integrinas/metabolismo , Marcação por Isótopo , Melanoma/diagnóstico por imagem , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Compostos Organofosforados/sangue , Compostos Organofosforados/farmacocinética , Peptídeos Cíclicos/sangue , Peptídeos Cíclicos/farmacocinética , Tomografia por Emissão de Pósitrons
20.
Anal Chim Acta ; 730: 66-70, 2012 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-22632046

RESUMO

The stability and in vivo robustness of [(177)Lu]Lu-DOTP as a potential bone-targeting radiopharmaceutical was determined with the aid of thermodynamic blood plasma modeling simulations. Glass electrode potentiometry was employed to measure the stability constants of the complexes of Lu(3+) with DOTP. Similarly, the complexes of DOTP with a selection of the important physiological metal ions: Ca(2+), Mg(2+), and Cu(2+) were determined, representing the typical interactions that the ligand would encounter upon administration. This made possible the construction of a blood plasma model of DOTP, aiding in establishing the potential susceptibility of the radiopharmaceutical. The ligand binds predominantly to calcium in vivo, accounting for 59.6% of that initially introduced as a component of the Lu-DOTP complex. Furthermore, due to a preference of the DOTP to bind to Cu(2+) it causes mobilization of the ions in blood plasma, and would therefore indicate a deficiency if the ligand is administered at a concentration of 8.5 × 10(-5) mol dm(-3). The lutetium-ions are preferentially bound to DOTP, with as much as 98.1% of the Lu(3+) occupying the ligand under physiological conditions.


Assuntos
Osso e Ossos/metabolismo , Complexos de Coordenação/metabolismo , Compostos Organometálicos/metabolismo , Compostos Radiofarmacêuticos/metabolismo , Neoplasias Ósseas/radioterapia , Cálcio/sangue , Cálcio/metabolismo , Complexos de Coordenação/sangue , Complexos de Coordenação/química , Cobre/sangue , Cobre/metabolismo , Estabilidade de Medicamentos , Humanos , Ligantes , Magnésio/sangue , Magnésio/metabolismo , Modelos Biológicos , Compostos Organometálicos/sangue , Compostos Organometálicos/química , Dor/radioterapia , Potenciometria , Compostos Radiofarmacêuticos/sangue , Compostos Radiofarmacêuticos/química , Termodinâmica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA