Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 250
Filtrar
1.
J Pharmacol Toxicol Methods ; 128: 107535, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38955285

RESUMEN

Quantification of the unbound portion of platinum (Pt) in human plasma is important for assessing the pharmacokinetics of the chemotherapeutic drug cisplatin. In this study, we sought to compare the recovery of unbound Pt using Nanosep® filters to 1) traditional filters (Centrifree®, Centrisart®, Amicon®) or trichloroacetic acid (TCA) protein precipitation, and 2) unbound, bound, and total Pt concentrations in clinical specimens. For the tested filters, the impact of 1) molecular weight cut-offs, 2) centrifugation force, and 3) total Pt concentration on Pt binding in human plasma was evaluated. Pt was quantified using inductively coupled-plasma mass spectrometry. In human plasma spiked with 0.9 µg/mL Pt, the percent of unbound Pt increased at higher centrifugation speeds. By comparison, the percent of unbound Pt was highest (42.1%) following TCA protein precipitation. When total Pt was ≤0.9 µg/mL, unbound Pt (∼20-30%) was consistent across filters. Conversely, when plasma was spiked with Pt exceeding 0.9 µg/mL, the percent of unbound Pt increased from 36.5 to 48% using ultrafiltration, compared to 63.4% to 79% with TCA precipitation. In patients receiving cisplatin-containing chemotherapy, the fraction of unbound Pt at concentrations exceeding 0.9 µg/mL ranged between 35 and 90%. Moreover, the unbound fraction of Pt in plasma correlated with the concentration of unbound (R2 = 0.738) and total Pt (R2 = 0.335). In summary, this study demonstrates that 1) the percent of unbound Pt is influenced by total and unbound Pt levels in vitro and in clinical specimens, and 2) ultrafiltration with Nanosep® filters is a feasible method for quantifying unbound Pt concentrations in human plasma.


Asunto(s)
Precipitación Química , Cisplatino , Ultrafiltración , Humanos , Ultrafiltración/métodos , Cisplatino/sangre , Cisplatino/farmacocinética , Platino (Metal)/sangre , Platino (Metal)/farmacocinética , Antineoplásicos/sangre , Antineoplásicos/farmacocinética , Unión Proteica , Ácido Tricloroacético/sangre
2.
Nucl Med Commun ; 43(11): 1121-1127, 2022 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-36120823

RESUMEN

OBJECTIVE: The platinum-based antineoplastic drug cisplatin is commonly used for chemotherapy in clinics. This work aims to demonstrate a radio-platinum tracer is useful for precisely quantifying small amounts of platinum in pharmacokinetics studies. METHODS: A cisplatin radiotracer (radio-cisplatin) was synthesized, and a comprehensive evaluation of cisplatin over 7 days after its intravenous injection into nude mice bearing a subcutaneous lung tumor (H460) was conducted. RESULTS: A biphasic retention curve in the whole body and blood was observed [ T1/2 (α) = 1.14 h, T1/2 (ß) = 5.33 days for the whole body, and T1/2 (α) = 23.9 min, T1/2 (ß) = 4.72 days for blood]. The blood concentration decreased within 1 day after injection. Most of the intact cisplatin was excreted via the kidneys in the early time points, and a small part was distributed in tissues including tumors. The plasma protein binding rate of cisplatin increased rapidly after injection, and the protein-bound cisplatin remained in the blood longer than intact cisplatin. The peak uptake in H460 tumors was 4.7% injected dose per gram at 15 min after injection, and the area under the curve (AUC 0-7 days ) was approximately one-half to one-third of the AUC 0-7 days in the kidneys, liver, and bone, where some toxicity is observed in humans. CONCLUSION: The radio-platinum tracer revealed the highly quantitative biodistribution of cisplatin, providing insights into the properties of cisplatin, including its adverse effects. The tracer enables a precise evaluation of pharmacokinetics for platinum-based drugs with high sensitivity.


Asunto(s)
Antineoplásicos , Neoplasias Pulmonares , Animales , Cisplatino , Humanos , Ratones , Ratones Desnudos , Platino (Metal)/farmacocinética , Distribución Tisular
3.
J Inorg Biochem ; 226: 111653, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34740039

RESUMEN

Theranostic anticancer agents with dual functions of diagnosis and therapy are in highly demand for breast cancer. Herein, a triphenylphosphonium (TPP)-decorated aggregation-induced emission (AIE)-based Pt(IV) prodrug ACPt was developed, which exhibited superior anticancer performance with novel anticancer mechanism of dual modulation of apoptosis and autophagy inhibition. The experimental data showed that ACPt induced increased reactive oxygen species (ROS), and decreased mitochondrial membrane potential (MMP). The morphology and function of mitochondria were also severely damaged and ACPt showed strong inhibition to both mitochondrial and glycolytic bioenergetics. Moreover, DNA damage and cell cycle arrest in the S-phase were also observed after the ACPt treatment, eventually leading to the apoptosis and autophagy inhibition of cancer cells. Furthermore, ACPt also indicated excellent anti-proliferation activity in 3D multicellular tumor spheroids (MCTSs), suggesting the potential to inhibit solid tumors in vivo. Our observation demonstrated that ACPt could serve as a promising anticancer theranostic agent toward breast cancers for prodrug activation monitoring and image-guided chemotherapy.


Asunto(s)
Antineoplásicos , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Neoplasias de la Mama , Complejos de Coordinación , Sistemas de Liberación de Medicamentos , Mitocondrias/metabolismo , Platino (Metal) , Profármacos , Células A549 , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Complejos de Coordinación/química , Complejos de Coordinación/farmacocinética , Complejos de Coordinación/farmacología , Femenino , Células Hep G2 , Humanos , Células MCF-7 , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Platino (Metal)/química , Platino (Metal)/farmacocinética , Platino (Metal)/farmacología , Profármacos/química , Profármacos/farmacocinética , Profármacos/farmacología
4.
J Nanobiotechnology ; 19(1): 415, 2021 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-34895243

RESUMEN

BACKGROUND: Immunochemotherapy is a potent anti-tumor strategy, however, how to select therapeutic drugs to enhance the combined therapeutic effect still needs to be explored. METHODS AND RESULTS: Herein, a magnetic resonance nanoprobe (MnP@Lip) with STING (Stimulator of INterferon Genes) activation character was synthesized and co-administered with platinum-based chemotherapeutics for enhanced immunochemotherapy. MnP@Lip nanoparticles was prepared by simple fabrication process with good reproducibility, pH-sensitive drug release behavior and biocompatibility. In vitro experiments elucidated that Mn2+ can promote the polarization of M0 and/or M2 macrophages to M1 phenotype, and promote the maturation of BMDC cells. Upon Mn2+ treatment, the STING pathway was activated in tumor cells, mouse lung epithelial cells, and immune cells. More importantly, anti-tumor experiments in vivo proved that MnP@Lip combined with platinum-based chemotherapeutics increased T cells infiltration in the tumor microenvironment, and inhibited tumor growth in the orthotopic therapeutic and postoperative tumor models. CONCLUSIONS: This kind of therapeutic strategy that combined MnP@Lip nanoparticles with platinum-based chemotherapeutics may provide a novel insight for immunochemotherapy.


Asunto(s)
Antineoplásicos , Sondas Moleculares , Nanopartículas , Platino (Metal) , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Línea Celular Tumoral , Células Cultivadas , Femenino , Inmunoterapia , Macrófagos/efectos de los fármacos , Imagen por Resonancia Magnética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Sondas Moleculares/química , Sondas Moleculares/farmacocinética , Nanopartículas/química , Nanopartículas/metabolismo , Neoplasias Experimentales , Platino (Metal)/química , Platino (Metal)/farmacocinética , Platino (Metal)/farmacología , Microambiente Tumoral/efectos de los fármacos
5.
ACS Appl Mater Interfaces ; 13(37): 44028-44040, 2021 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-34499483

RESUMEN

Polymeric nanocapsules hold considerable applications in cancer drug delivery, but the synthesis of well-defined nanocapsules with a tunable drug release property remains a significant challenge in fabrication. Herein, we demonstrate a supramolecular complexation strategy to assemble small molecular platinum (Pt) compounds into well-defined nanocapsules with high drug loading, acidity-sensitivity, and tunable Pt releasing profile. The design utilizes poly(ethylene glycol)-dendritic polylysine-G4/amides to complex with Pt compounds, forming stable nanocapsules with diameters approximately ∼20 nm and membrane thickness around several nanometers. The stability, drug content, and release profiles are tunable by tailoring the dendritic structure. The designated polymer-Pt nanocapsules, PEG-G4/MSA-Pt, showed sustained blood retention, preferential tumor accumulation, enhanced cellular uptake, lysosomal drug release, and nuclear delivery capability. PEG-G4/MSA-Pt showed enhanced antitumor efficacy compared to free cisplatin and other nanocapsules, which stopped the progression of both A549 cell xenografts and patient-derived xenografts (PDXs) of hepatocellular carcinoma on a mice tumor model. Thus, we believe this strategy is promising for developing Pt-based nanomedicine for cancer drug delivery.


Asunto(s)
Antineoplásicos/uso terapéutico , Cisplatino/uso terapéutico , Portadores de Fármacos/química , Nanocápsulas/química , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Cisplatino/química , Cisplatino/farmacocinética , Complejos de Coordinación/química , Complejos de Coordinación/farmacocinética , Dendrímeros/química , Dendrímeros/farmacocinética , Portadores de Fármacos/farmacocinética , Liberación de Fármacos , Humanos , Ratones Endogámicos BALB C , Platino (Metal)/química , Platino (Metal)/farmacocinética , Polietilenglicoles/química , Polietilenglicoles/farmacocinética , Polilisina/química , Polilisina/farmacocinética , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Int J Mol Sci ; 22(11)2021 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-34070401

RESUMEN

Transition metal coordination compounds play an important role in the treatment of neoplastic diseases. However, due to their low selectivity and bioavailability, as well as the frequently occurring phenomenon of drug resistance, new chemical compounds that could overcome these phenomena are still being sought. The solution seems to be the synthesis of new metal complexes conjugated with drug carriers, e.g., dendrimers. Numerous literature data have shown that dendrimers improve the bioavailability of the obtained metal complexes, solving the problem of their poor solubility and stability in an aqueous environment and also breaking down inborn and acquired drug resistance. Therefore, the aim of this study was to synthesize a novel imidazole platinum(II) complex conjugated with and without the second-generation PAMAM dendrimer (PtMet2-PAMAM and PtMet2, respectively) and to evaluate its antitumor activity. Cell viability studies indicated that PtMet2-PAMAM exhibited higher cytotoxic activity than PtMet2 in MCF-7 and MDA-MB-231 breast cancer cells at relatively low concentrations. Moreover, our results indicated that PtMet2-PAMAM exerted antiproliferative effects in a zebrafish embryo model. Treatment with PtMet2-PAMAM substantially increased apoptosis in a dose-dependent manner via caspase-9 (intrinsic pathway) and caspase-8 (extrinsic pathway) activation along with pro-apoptotic protein expression modulation. Additionally, we showed that apoptosis can be induced by activating POX, which induces ROS production. Furthermore, our results also clearly showed that the tested compounds trigger autophagy through p38 pathway activation and increase Beclin-1, LC3, AMPK, and mTOR inhibition. The high pro-apoptotic activity and the ability to activate autophagy by the imidazole platinum(II) complex conjugated with a dendrimer may be due to its demonstrated ability to reverse multidrug resistance (MDR) and thereby increase cellular accumulation in breast cancer cells.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Complejos de Coordinación , Dendrímeros , Imidazoles , Platino (Metal) , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Complejos de Coordinación/síntesis química , Complejos de Coordinación/química , Complejos de Coordinación/farmacocinética , Complejos de Coordinación/farmacología , Dendrímeros/química , Dendrímeros/farmacocinética , Dendrímeros/farmacología , Femenino , Humanos , Imidazoles/química , Imidazoles/farmacocinética , Imidazoles/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Células MCF-7 , Proteínas de Neoplasias/metabolismo , Platino (Metal)/química , Platino (Metal)/farmacocinética , Platino (Metal)/farmacología
7.
Vet Surg ; 49(4): 748-757, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-31944331

RESUMEN

OBJECTIVE: To evaluate the pharmacokinetics (PK) of platinum (Pt) and safety of carboplatin-impregnated calcium sulfate hemihydrate (C-I CSH) beads after implantation in healthy cats. STUDY DESIGN: In vivo experimental study. ANIMALS: Six healthy adult cats. METHODS: Three C-I CSH beads were implanted in muscle pockets over the right and left hemithoraces of each cat (~3.9 mg/kg of Pt; 60.4 mg/m2 of calculated carboplatin). Hematology and blood chemistry were tested at baseline and 3, 7, 14, and 21 days postimplantation. Serum was analyzed for Pt at specific times from 1 hour to 21 days. Tissue was obtained for histopathology and analysis of Pt at 3, 7, 14, and 21 days at standardized distances from implantation sites. RESULTS: Platinum was detected in tissues at all times and distances (range, 0.1-4.19 µg/g). Serum Pt increased up to 2.6 hours (3.25 µg/mL) then decreased sharply. Samples containing muscle had higher Pt compared with samples without muscle (P = .004). Mild hypercalcemia was noted in four cats, and mild inflammatory reaction was noted on histopathology of all samples. CONCLUSION: Platinum was released from C-I CSH beads differentially into surrounding tissues over 21 days. Systemic absorption of Pt was minimal, but mild hypercalcemia occurred. CLINICAL SIGNIFICANCE: Implantation was well tolerated by healthy adult cats. Securing beads within muscle may limit Pt diffusion to targeted tissue. Although Pt concentrations did not achieve levels reported to be cytotoxic for feline sarcoma cells in culture, results provide evidence to support evaluation of efficacy in the tumor microenvironment of cats with locally invasive cancers.


Asunto(s)
Antineoplásicos/efectos adversos , Sulfato de Calcio/efectos adversos , Carboplatino/efectos adversos , Platino (Metal)/farmacocinética , Animales , Gatos , Femenino
8.
Chemosphere ; 242: 124967, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31677506

RESUMEN

Mechanistic models based on chemical properties of metals and body size have received substantial attention for their potential application to various metals and to different conditions without required calibration. This advantage has been demonstrated for a number of metals, such as Cd and Ag. However, the capacity of metal-specific chemical properties to explain variations in the accumulation for platinum-group elements (PGEs) has not been investigated yet, although emission of these metals is of increasing concern. Once being released, PGEs exist in the environment in mixtures with other metals. The present study attempted to model the accumulation of Pd and Pt in mixtures with Ag and Cd in the zebra mussel (Dreissena polymorpha) from the aqueous phase; and to investigate the potential application of mechanistic models to Pd and Pt. The present study showed statistically insignificant differences in metal accumulation among size groups in a narrow range of shell length (16-22 mm). Kinetic models could simulate well the accumulation of Cd, Ag, and Pt when metal-specific responses of zebra mussels are taken into consideration. These responses include enhanced immobilisation as a detoxifying mechanism and exchange between soft tissues and shells via the extrapallial fluid. Environmental conditions, e.g. the presence of abiotic ligands such as chloride, might also play an important role in metal accumulation. Significant relationships between the absorption efficiency and the covalent index indicate the potential application of mechanistic models based on this chemical property to Pt.


Asunto(s)
Dreissena/metabolismo , Modelos Químicos , Contaminantes Químicos del Agua/farmacocinética , Animales , Bioacumulación , Cadmio/farmacocinética , Cinética , Paladio/farmacocinética , Platino (Metal)/farmacocinética , Plata/farmacocinética , Contaminantes Químicos del Agua/análisis
9.
Med Pr ; 70(4): 487-495, 2019 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-31162484

RESUMEN

Platinum nanoparticles (PtNPs) have been widely used not only in industry, but above all in medicine and diagnostics. However, there are disturbing reports related to the toxic effects of nanoplatinum, which is the main reason why the authors of this study have decided to review and analyze literature data related to its toxicity and impact on human health. While PtNPs may be absorbed by the respiratory and digestive tract, and can penetrate through the epidermis, there is no evidence concerning their absorption through the skin. Platinum nanoparticles accumulate mainly in the liver and spleen although they also reach other internal organs, such as lungs, kidneys or heart. Toxicokinetics of platinum nanoparticles depends strongly on the particle size. Only few studies regarding platinum nanoparticles toxicity have been conducted. Animals intratracheally exposed to platinum nanoparticles have demonstrated an increased level of proinflammatory cytokines in bronchoalveolar lavage which confirms inflammatory response in the lungs. Oral administration of PtNPs can cause inflammatory response and induce oxidative stress. Nanoplatinum has been found to induce hepatotoxicity and nephrotoxicity via the intravenous route. It can cause DNA damage and cellular apoptosis without significant cytotoxicity. There are no research studies on its carcinogenicity. Fetal or maternal toxicity has not been observed, but an increased mortality and a decreased growth of the offspring have been demonstrated. Platinum nanoparticles may permeate the skin barrier but there is no evidence for their absorption. Due to the insufficient number of tests that have been carried out to date, it is not possible to clearly determine the occupational exposure limit value; however, caution is recommended to employees exposed to their effects. Med Pr. 2019;70(4):487-95.


Asunto(s)
Nanopartículas del Metal/toxicidad , Platino (Metal)/toxicidad , Animales , Humanos , Inflamación , Nanopartículas del Metal/química , Exposición Profesional , Estrés Oxidativo , Tamaño de la Partícula , Platino (Metal)/farmacocinética
10.
ACS Nano ; 13(6): 6647-6661, 2019 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-31083971

RESUMEN

Highly efficient nanoarchitectures are of great interest for achieving precise chemotherapy with minimized adverse side effects in cancer therapy. However, a major challenge remains in exploring a rational approach to synthesize spatiotemporally selective vehicles for precise cancer chemotherapy. Here, we demonstrate a rational design of bifunctional light-activatable platinum nanocomplexes (PtNCs) that produce dually cooperative cancer therapy through spatiotemporally selective thermo-chemotherapy. The Pt4+-coordinated polycarboxylic nanogel is explored as the nanoreactor template, which is exploited to synthesize bifunctional PtNCs consisting of a zero-valent Pt0 core and a surrounding bivalent Pt2+ shell with tunable ratios through a facile and controllable reduction. Without light exposure, chemotherapeutic Pt2+ ions are tightly bound on the surface of PtNCs, efficiently reducing undesirable drug leakage and nonselective damage on normal tissues/cells. Upon light exposure, PtNCs generate much heat via photothermal conversion from the Pt0 core and simultaneously trigger a rapid release of chemotherapeutic Pt2+ ions, thereby leading to the spatiotemporally light-activatable synergistic effect of thermo-chemotherapy. Moreover, PtNCs show enhanced tumor accumulation through the heat-triggered hydrophilicity-hydrophobicity transition upon immediate light exposure after injection, dramatically facilitating in vivo tumor regression through their cooperative anticancer efficiency. This rational design of spatiotemporally activatable nanoparticles provides an insightful tool for precise cancer therapy.


Asunto(s)
Antineoplásicos/administración & dosificación , Liberación de Fármacos , Nanoconjugados/química , Neoplasias Experimentales/tratamiento farmacológico , Fotoquimioterapia/métodos , Platino (Metal)/administración & dosificación , Células 3T3 , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Células Hep G2 , Humanos , Luz , Ratones , Ratones Endogámicos BALB C , Nanoconjugados/efectos de la radiación , Platino (Metal)/farmacocinética , Platino (Metal)/uso terapéutico , Distribución Tisular
11.
Anal Chem ; 90(15): 8831-8837, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-29961333

RESUMEN

Laterally resolved chemical analysis (chemical imaging) has increasingly attracted attention in the Life Sciences during the past years. While some developments have provided improvements in lateral resolution and speed of analysis, there is a trend toward the combination of two or more analysis techniques, so-called multisensor imaging, for providing deeper information into the biochemical processes within one sample. In this work, a human malignant pleural mesothelioma sample from a patient treated with cisplatin as a cytostatic agent has been analyzed using laser ablation inductively coupled plasma mass spectrometry (LA-ICPMS) and matrix-assisted laser desorption/ionization mass spectrometry (MALDI MS). While LA-ICPMS was able to provide quantitative information on the platinum distribution along with the distribution of other elemental analytes in the tissue sample, MALDI MS could reveal full information on lipid distributions, as both modes of polarity, negative and positive, were used for measurements. Tandem MS experiments verified the occurrence of distinct lipid classes. All imaging analyses were performed using a lateral resolution of 40 µm, providing information with excellent depth of details. By analyzing the very same tissue section, it was possible to perfectly correlate the obtained analyte distribution information in an evaluation approach comprising LA-ICPMS and MALDI MS data. Correlations between platinum, phosphorus, and lipid distributions were found by the use of advanced statistics. The present proof-of-principle study demonstrates the benefit of data combination for outcomes beyond one method imaging modality and highlights the value of advanced chemical imaging in the Life Sciences.


Asunto(s)
Lípidos/análisis , Neoplasias Pulmonares/química , Mesotelioma/química , Fósforo/análisis , Platino (Metal)/análisis , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Antineoplásicos/análisis , Antineoplásicos/farmacocinética , Cisplatino/análisis , Cisplatino/farmacocinética , Cisplatino/uso terapéutico , Elementos Químicos , Humanos , Terapia por Láser , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Mesotelioma/diagnóstico por imagen , Mesotelioma/tratamiento farmacológico , Mesotelioma/patología , Mesotelioma Maligno , Imagen Molecular/métodos , Imagen Multimodal/métodos , Análisis Multivariante , Platino (Metal)/farmacocinética , Platino (Metal)/uso terapéutico , Pleura/química , Pleura/diagnóstico por imagen , Pleura/efectos de los fármacos , Pleura/patología , Manejo de Especímenes , Espectrometría de Masas en Tándem/métodos
12.
Toxicol Lett ; 290: 73-82, 2018 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-29574132

RESUMEN

Cisplatin (CP) is a widely used drug in treatment of solid tumors. However, the use of CP was hampered by its serious side effects especially nephrotoxicity. This study aims to investigate the effect of resveratrol (RES) on CP-induced nephrotoxicity, particularly, the effect of RES on CP pharmacokinetics (PKs). Male white albino rats were divided to four group's six rats each. The first group received (1%) tween 80 in normal saline and served as control. The second group received RES (30 mg kg-1) per day for 14 consecutive day's i.p. The third and fourth groups were given a single i.p. injection of CP (6 mg kg-1) with or without pre-treatment of RES (30 mg kg-1per day for 14 consecutive days), respectively. Following administration of CP, plasma, urine and kidney platinum concentration were monitored to study PKs of CP. Five days after the CP injection, rats were killed; blood samples were collected; kidneys were dissected; and biochemical, immunohistochemical, and histological examinations were performed. Our results revealed that CP treatment significantly deteriorated kidney functions with subsequent alteration in redox balance of the kidney. On the other hand, RES successfully ameliorated CP-induced kidney injury and recovered normal kidney tissue redox status. Importantly, while RES pre-treatment did not significantly alter the plasma CP level, it dramatically decreased the urine concentration of CP and lowered its accumulation into the kidneys. Moreover, it increased CP plasma half-life (t1/2) with subsequent decrease in its elimination rate constant, indicating an important role of PKs modulation in RES protection against CP-induced renal damage. Taken together, RES may protect the kidney tissue from the deleterious effects of CP through constringe of CP renal accumulation and enhancement of CP-induced oxidative stress.


Asunto(s)
Antineoplásicos/toxicidad , Cisplatino/toxicidad , Riñón/efectos de los fármacos , Platino (Metal)/farmacocinética , Estilbenos/farmacología , Amidinotransferasas/genética , Animales , Cisplatino/farmacocinética , Riñón/metabolismo , Riñón/patología , Masculino , Óxido Nítrico Sintasa de Tipo III/genética , Ratas , Ratas Wistar , Resveratrol
13.
Biomaterials ; 154: 248-260, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29144983

RESUMEN

Photoactive noble metal nanoparticles are of increasing importance toward personalized cancer therapy in the field of precision nanomedicine. A critical challenge remains in the exploration of clinically potential noble metal nanoparticles for highly efficient cancer theranostics. Here, we introduce albumin-coordinated assembly of clearable Pt nanodots (Pt-NDs) with monodisperse nanostructure as high-performance theranostic agents for imaging-guided photothermal tumor ablation. We precisely manipulate the reduction and growth of tetravalent Pt ions into ultrasmall nanodots through albumin-directed growth kinetics, thereby leading to the synthesis of monodisperse 6.7 nm Pt-NDs with albumin molecules as the corona. Pt-NDs exhibit the surface plasmon resonance at 225 nm with enhanced near-infrared (NIR) absorbance, ideal resistance to photo-bleaching, distinct photoacoustic and X-ray signals, as well as remarkable photothermal effect through non-radiative relaxation under NIR light irradiation. In particular, Pt-NDs possess preferable tumor accumulation, and effective in vivo excretory capability. Thus, these nanodots promote preferable in vivo microscopic photoacoustics and spatially anatomic CT imaging with enhanced contrast, as well as potent hyperthermia-mediated tumor ablation. These findings represent a facile and general approach to fabricate high-performance noble metal nanostructures with clinical potential for cancer theranostics.


Asunto(s)
Albúminas/química , Nanoestructuras/química , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Fototerapia , Platino (Metal)/uso terapéutico , Nanomedicina Teranóstica , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Endocitosis/efectos de los fármacos , Fluorescencia , Humanos , Cinética , Ratones Endogámicos BALB C , Ratones Desnudos , Imagen Multimodal , Nanoestructuras/ultraestructura , Neoplasias/patología , Técnicas Fotoacústicas , Platino (Metal)/farmacocinética , Platino (Metal)/farmacología , Distribución Tisular/efectos de los fármacos , Tomografía Computarizada por Rayos X
14.
J Inorg Biochem ; 176: 175-180, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28917640

RESUMEN

Three biotinylated platinum(IV) complexes (1-3) were designed and synthesized. The resulting platinum(IV) complexes exhibited effective cytotoxicity against the tested cancer cell lines, especially complex 1, which was 2.0-9.6-fold more potent than cisplatin. These complexes were found to be rapidly reduced to their activated platinum(II) counterparts by glutathione or ascorbic acid under biologically relevant condition. Additional molecular docking studies revealed that the biotin moieties of all Pt(IV) complexes can effectively bind with the streptavidin through the noncovalent interactions. Besides, introduction of the biotin group can obviously promote the cancer cell uptake of platinum when treated with complex 1, particularly in cisplatin-resistant SGC-7901/Cis cancer cells. Further mechanistic studies on complex 1 indicated that it activated the expression of Bax, and induced cytochrome c release from the mitochondria, and finally activated caspase-3.


Asunto(s)
Antineoplásicos , Biotina , Complejos de Coordinación , Sistemas de Liberación de Medicamentos/métodos , Neoplasias/tratamiento farmacológico , Platino (Metal) , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Biotina/química , Biotina/farmacocinética , Biotina/farmacología , Biotinilación , Complejos de Coordinación/síntesis química , Complejos de Coordinación/química , Complejos de Coordinación/farmacocinética , Complejos de Coordinación/farmacología , Células Hep G2 , Humanos , Células MCF-7 , Simulación del Acoplamiento Molecular , Neoplasias/metabolismo , Platino (Metal)/química , Platino (Metal)/farmacocinética , Platino (Metal)/farmacología
15.
Neuroreport ; 28(9): 506-513, 2017 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-28471848

RESUMEN

The blood-brain barrier (BBB) protects the central nervous system from external insults by limiting substance diffusion through the endothelial interface. The presence of the BBB makes drug delivery in neurological disorders very challenging. Cisplatin has been shown to be cytotoxic to glioma cells, but substantial limitations exist in its clinical applications due to difficulties in penetration across the BBB. Here, we show that L-borneol, a messenger drug widely used in traditional Chinese medicine, can induce transient disruption of the BBB after 20 min of oral administration. The permeability of the BBB began to recover within 1 h of the administration of L-borneol. Different dosages of L-borneol (100, 150, 300, 600, and 900 mg/kg) could induce significant Evans blue leakage (P<0.05). Oral administration of L-borneol elevated cisplatin concentrations in peritumoral tissue (1.24±0.12 µg/g) and tumor loci (1.41±0.13 µg/g), compared with those in the paraffin control (0.88±0.10 and 0.92±0.15 µg/g, respectively) (P<0.05). Furthermore, we found that the median survival period of tumor-bearing mice was significantly higher in the cisplatin plus L-borneol group (24.0±4.9 days) than in the cisplatin plus vehicle group (19.3±3.9 days) (P<0.05). The neurological deficits were more severe in the vehicle and cisplatin plus vehicle groups at 14 and 21 days after implantation of intracranial glioma cells than in the cisplatin plus L-borneol group. In conclusion, our results indicate that the transient opening of the BBB induced by L-borneol could enhance cisplatin accumulation within the glioma tissue and improve the survival of tumor-bearing mice.


Asunto(s)
Antineoplásicos/uso terapéutico , Barrera Hematoencefálica/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Canfanos/farmacología , Cisplatino/uso terapéutico , Glioma/tratamiento farmacológico , Animales , Barrera Hematoencefálica/fisiología , Neoplasias Encefálicas/complicaciones , Neoplasias Encefálicas/diagnóstico por imagen , Canfanos/uso terapéutico , Permeabilidad Capilar/efectos de los fármacos , Claudina-5/metabolismo , Medios de Contraste/farmacocinética , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Glioma/complicaciones , Glioma/diagnóstico por imagen , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Enfermedades del Sistema Nervioso/etiología , Ocludina/metabolismo , Platino (Metal)/farmacocinética
16.
J Biol Inorg Chem ; 22(5): 765-774, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28516214

RESUMEN

There is much interest in understanding the mechanisms by which platinum-based anticancer agents enter cells, and the copper transporter CTR1 has been the focus of many recent studies. While there is a clinical correlation between CTR1 levels and platinum efficacy, cellular studies have provided conflicting evidence relating to the relationship between cisplatin and CTR1. We report here our studies of the relationship between cisplatin and copper homeostasis in human colon cancer cells. While the accumulation of copper and platinum do not appear to compete with each other, we did observe that cisplatin perturbs CTR1 distribution within 10 min, a far shorter incubation time than commonly employed in cellular studies of cisplatin. Furthermore, on these short time-scales, cisplatin caused an increase in the cytoplasmic labile copper pool. While the predominant focus of studies to date has been on CTR1, these studies highlight the importance of investigating the interaction of cisplatin with other copper proteins.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Cisplatino/metabolismo , Neoplasias del Colon/metabolismo , Proteínas de Transporte de Catión/química , Supervivencia Celular , Cisplatino/química , Neoplasias del Colon/patología , Cobre/metabolismo , Cobre/farmacocinética , Transportador de Cobre 1 , Relación Dosis-Respuesta a Droga , Homeostasis , Humanos , Platino (Metal)/metabolismo , Platino (Metal)/farmacocinética , Relación Estructura-Actividad
17.
Eur J Nucl Med Mol Imaging ; 44(8): 1347-1354, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28190123

RESUMEN

AIMS: In vivo biodistribution imaging of platinum-based compounds may allow better patient selection for treatment with chemo(radio)therapy. Radiolabeling with Platinum-195m (195mPt) allows SPECT imaging, without altering the chemical structure or biological activity of the compound. We have assessed the feasibility of 195mPt SPECT imaging in mice, with the aim to determine the image quality and accuracy of quantification for current preclinical imaging equipment. METHODS: Enriched (>96%) 194Pt was irradiated in the High Flux Reactor (HFR) in Petten, The Netherlands (NRG). A 0.05 M HCl 195mPt-solution with a specific activity of 33 MBq/mg was obtained. Image quality was assessed for the NanoSPECT/CT (Bioscan Inc., Washington DC, USA) and U-SPECT+/CT (MILabs BV, Utrecht, the Netherlands) scanners. A radioactivity-filled rod phantom (rod diameter 0.85-1.7 mm) filled with 1 MBq 195mPt was scanned with different acquisition durations (10-120 min). Four healthy mice were injected intravenously with 3-4 MBq 195mPt. Mouse images were acquired with the NanoSPECT for 120 min at 0, 2, 4, or 24 h after injection. Organs were delineated to quantify 195mPt concentrations. Immediately after scanning, the mice were sacrificed, and the platinum concentration was determined in organs using a gamma counter and graphite furnace - atomic absorption spectroscopy (GF-AAS) as reference standards. RESULTS: A 30-min acquisition of the phantom provided visually adequate image quality for both scanners. The smallest visible rods were 0.95 mm in diameter on the NanoSPECT and 0.85 mm in diameter on the U-SPECT+. The image quality in mice was visually adequate. Uptake was seen in the kidneys with excretion to the bladder, and in the liver, blood, and intestine. No uptake was seen in the brain. The Spearman correlation between SPECT and gamma counter was 0.92, between SPECT and GF-AAS it was 0.84, and between GF-AAS and gamma counter it was0.97 (all p < 0.0001). CONCLUSION: Preclinical 195mPt SPECT is feasible with acceptable tracer doses and acquisition times, and provides good image quality and accurate signal quantification.


Asunto(s)
Platino (Metal) , Tomografía Computarizada por Tomografía Computarizada de Emisión de Fotón Único/métodos , Animales , Ratones , Fantasmas de Imagen , Platino (Metal)/química , Platino (Metal)/farmacocinética , Radioquímica , Distribución Tisular
18.
Arch Toxicol ; 91(2): 785-797, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27307157

RESUMEN

The therapeutic efficacy of the anticancer drug cisplatin is limited by the development of resistance. We therefore investigated newly synthesized platinum-nitroxyl complexes (PNCs) for their potential to circumvent cisplatin resistance. The complexes used were PNCs with bivalent cis-PtII(R·NH2)(NH3)Cl2 and cis-PtII(DAPO)Ox and four-valent platinum cis,trans,cis-PtIV(R·NH2)(NH3)(OR)2Cl2 and cis,trans,cis-PtIV(DAPO)(OR)2Ox, where R· are TEMPO or proxyl nitroxyl radicals, DAPO is trans-3,4-diamino-2,2,6,6-tetramethylpiperidine-1-oxyl, and OR and Ox are carboxylato and oxalato ligands, respectively. The complexes were characterized by spectroscopic methods, HPLC, log P ow data and elemental analysis. We studied intracellular platinum accumulation, DNA platination and cytotoxicity upon treatment with the PNCs in a model system of the bladder cancer cell line RT112 and its cisplatin-resistant subline RT112-CP. Platinum accumulation and DNA platination were similar in RT112 and RT112-CP cells for both bivalent and four-valent PNCs, in contrast to cisplatin for which a reduction in intracellular accumulation and DNA platination was observed in the resistant subline. The PNCs were found to platinate DNA in relation to the length of their axial RO-ligands. Furthermore, the PNCs were increasingly toxic in relation to the elongation of their axial RO-ligands, with similar toxicities in RT112 and its cisplatin-resistant subline. Using a cell-free assay, we observed induction of oxidative DNA damage by cisplatin but not PNCs suggesting that cisplatin exerts its toxic action by platination and oxidative DNA damage, while cells treated with PNCs are protected against oxidatively induced lesions. Altogether, our study suggests that PNCs may provide a more effective treatment for tumors which have developed resistance toward cisplatin.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Compuestos de Platino/farmacología , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Antineoplásicos/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Técnicas de Química Sintética , ADN/química , Roturas del ADN/efectos de los fármacos , Humanos , Óxidos de Nitrógeno/química , Platino (Metal)/química , Platino (Metal)/farmacocinética , Compuestos de Platino/química , Neoplasias de la Vejiga Urinaria/patología
19.
J Biochem Mol Toxicol ; 31(1): 1-9, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27550472

RESUMEN

Cisplatin (CP) is one of the most effective chemotherapeutic agents. Unfortunately, CP-induced nephrotoxicity hampered its use. This study aims to investigate the effect of vitamin E (Vit E) on CP-induced nephrotoxicity. Male white albino rats were divided to four group's six rats each and received either, 1% tween 80 in normal saline or Vit E (75 mg/kg) per day for 14 consecutive days or a single injection of CP (6 mg/kg) alone or CP (6 mg/kg) together with Vit E (75 mg/kg per day for 14 consecutive days). Five days after the CP injection, rats were euthanized; blood samples were collected; kidneys were dissected; and biochemical, immunohistochemical, and histological examinations were performed. Our results revealed that CP treatment significantly increased serum levels of creatinine and urea. Moreover, reduced glutathione (GSH) content as well as superoxide dismutase (SOD) and catalase (CAT) activities were significantly reduced with concurrent increase in kidney malondialdehyde (MDA) content following CP treatment. Vit E successfully lowered serum levels of urea and creatinine, enhanced creatinine clearance and diuresis, and normalized relative kidney/body weight. Furthermore, Vit E successfully normalized renal MDA and nitrite concentrations, elevated GSH level, and restored CAT and SOD activities in renal tissues. Histopathological examination of rat kidney revealed that Vit E significantly mitigated CP-induced renal damage. Importantly, administration of Vit E reduced kidney total platinum concentration indicating a role of platinum renal accumulation on the ability of Vit E to protect against CP nephrotoxicity.


Asunto(s)
Cisplatino , Enfermedades Renales , Riñón , Estrés Oxidativo/efectos de los fármacos , Platino (Metal) , Vitamina E/farmacología , Animales , Cisplatino/efectos adversos , Cisplatino/farmacocinética , Cisplatino/farmacología , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/patología , Inflamación/prevención & control , Riñón/metabolismo , Riñón/patología , Enfermedades Renales/inducido químicamente , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Enfermedades Renales/prevención & control , Masculino , Nitrosación/efectos de los fármacos , Platino (Metal)/efectos adversos , Platino (Metal)/farmacocinética , Platino (Metal)/farmacología , Ratas , Ratas Wistar
20.
Mol Cancer Ther ; 16(2): 376-387, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27903751

RESUMEN

We report progress on predicting tumor response to platinum-based chemotherapy with a novel mass spectrometry approach. Fourteen bladder cancer patients were administered one diagnostic microdose each of [14C]carboplatin (1% of the therapeutic dose). Carboplatin-DNA adducts were quantified by accelerator mass spectrometry in blood and tumor samples collected within 24 hours, and compared with subsequent chemotherapy response. Patients with the highest adduct levels were responders, but not all responders had high adduct levels. Four patient-derived bladder cancer xenograft mouse models were used to test the possibility that another drug in the regimen could cause a response. The mice were dosed with [14C]carboplatin or [14C]gemcitabine and the resulting drug-DNA adduct levels were compared with tumor response to chemotherapy. At least one of the drugs had to induce high drug-DNA adduct levels or create a synergistic increase in overall adducts to prompt a corresponding therapeutic response, demonstrating proof-of-principle for drug-DNA adducts as predictive biomarkers. Mol Cancer Ther; 16(2); 376-87. ©2016 AACR.


Asunto(s)
Antineoplásicos/administración & dosificación , Biomarcadores , Aductos de ADN , Resistencia a Antineoplásicos , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Carboplatino/administración & dosificación , Carboplatino/efectos adversos , Carboplatino/sangre , Carboplatino/metabolismo , Carboplatino/farmacocinética , Línea Celular Tumoral , Aductos de ADN/sangre , Aductos de ADN/metabolismo , Reparación del ADN , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacocinética , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Femenino , Humanos , Espectrometría de Masas , Ratones , Mutación , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/mortalidad , Platino (Metal)/administración & dosificación , Platino (Metal)/efectos adversos , Platino (Metal)/farmacocinética , Neoplasias de la Vejiga Urinaria/diagnóstico , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA