Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 513
Filtrar
1.
Inorg Chem ; 61(7): 3240-3248, 2022 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-35137586

RESUMEN

Arsenoplatin-1 (AP-1) is a dual-action anticancer metallodrug with a promising pharmacological profile that features the simultaneous presence of a cisplatin-like center and an arsenite center. We investigated its interactions with proteins through a joint experimental and theoretical approach. The reactivity of AP-1 with a variety of proteins, including carbonic anhydrase (CA), superoxide dismutase (SOD), myoglobin (Mb), glyceraldehyde 3-phosphate dehydrogenase (GAPDH), and human serum albumin (HSA), was analyzed by means of electrospray ionization mass spectrometry (ESI MS) measurements. In accordance with previous observations, ESI MS experiments revealed that the obtained metallodrug-protein adducts originated from the binding of the [(AP-1)-Cl]+ fragment to accessible protein residues. Remarkably, in two cases, i.e., Mb and GAPDH, the formation of a bound metallic fragment that lacked the arsenic center was highlighted. The reactions of AP-1 with various nucleophiles side chains of neutral histidine, methionine, cysteine, and selenocysteine, in neutral form as well as cysteine and selenocysteine in anionic form, were subsequently analyzed through a computational approach. We found that the aquation of AP-1 is energetically disfavored, with a reaction free energy of +19.2 kcal/mol demonstrating that AP-1 presumably attacks its biological targets through the exchange of the chloride ligand. The theoretical analysis of thermodynamics and kinetics for the ligand-exchange processes of AP-1 with His, Met, Cys, Sec, Cys-, and Sec- side chain models unveils that only neutral histidine and deprotonated cysteine and selenocysteine are able to effectively replace the chloride ligand in AP-1.


Asunto(s)
Trióxido de Arsénico/análogos & derivados , Cisplatino/análogos & derivados
2.
J Mol Model ; 28(1): 2, 2021 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-34874466

RESUMEN

Cisplatin is a widely used anti-cancer drug which inhibits the replication and polymerization of DNA molecule while showing some side effects and drug resistance. For this reason, to enhance its therapeutic index, researchers have synthesized several thousand analogs and tested their properties. In this project, several cisplatin analogs were designed to theoretically study the biological activity and lipophilicity effects on amine changes. The amines of the cisplatin molecule were substituted with aliphatic amines in different analogs. Computational methods such as molecular dynamics simulation, molecular docking, and molecular mechanics Poisson-Boltzmann surface area analysis were performed to investigate the binding of six cisplatin derivatives with DNA. The binding affinity and potential interactions of these drugs with double-strand DNA were analyzed. The stability effect of these drugs was investigated via root-mean-square deviation and root-mean-square fluctuation analysis, which showed that some analogs can break base-pair interaction at the end of DNA and reduced the stability of DNA. Also, the results revealed that the hydrogen bond is one of the most important factors in the binding of cisplatin's adduct to DNA. Molecular mechanics Poisson-Boltzmann surface area analysis indicated that electrostatic and van der Waals interactions are the most important deriving forces to the binding of cisplatin's drug to DNA. Finally, data revealed that cisplatin and the cis-dichloro-dimethylamine-platin tendency for binding to DNA are greater than that of other analogs.


Asunto(s)
Aminas/química , Antineoplásicos/química , Cisplatino/química , ADN/química , Antineoplásicos/farmacología , Cisplatino/análogos & derivados , Cisplatino/farmacología , Humanos , Enlace de Hidrógeno , Ligandos , Conformación Molecular , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Estructura Molecular , Reproducibilidad de los Resultados , Relación Estructura-Actividad
3.
Int J Mol Sci ; 22(4)2021 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-33668605

RESUMEN

Arsenoplatin-1 (AP-1), the prototype of a novel class of metallodrugs containing a PtAs(OH)2 core, was encapsulated within the apoferritin (AFt) nanocage. UV-Vis absorption spectroscopy and inductively coupled plasma-atomic emission spectroscopy measurements confirmed metallodrug encapsulation and allowed us to determine the average amount of AP-1 trapped inside the cage. The X-ray structure of AP-1-encapsulated AFt was solved at 1.50 Å. Diffraction data revealed that an AP-1 fragment coordinates the side chain of a His residue. The biological activity of AP-1-loaded AFt was comparatively tested on a few representative cancer and non-cancer cell lines. Even though the presence of the cage reduces the overall cytotoxicity of AP-1, it improves its selectivity towards cancer cells.


Asunto(s)
Antineoplásicos , Trióxido de Arsénico/análogos & derivados , Cisplatino/análogos & derivados , Citotoxinas , Ferritinas , Neoplasias/tratamiento farmacológico , Compuestos de Platino , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Trióxido de Arsénico/química , Trióxido de Arsénico/farmacología , Células 3T3 BALB , Cisplatino/química , Cisplatino/farmacología , Citotoxinas/química , Citotoxinas/farmacología , Ferritinas/química , Ferritinas/farmacología , Humanos , Ratones , Estructura Molecular , Neoplasias/metabolismo , Neoplasias/patología , Compuestos de Platino/química , Compuestos de Platino/farmacología , Relación Estructura-Actividad
4.
Biochem Pharmacol ; 188: 114523, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33741331

RESUMEN

Triple-negative breast cancer (TNBC) shares the molecular features facilitating epithelial-to-mesenchymal transition (EMT), which contributed to tumor invasion and metastasis. A platinum(IV) conjugate ketoplatin deriving from FDA-approved drugs cisplatin and ketoprofen was designed and prepared to enhance antitumor activity and suppress EMT in TNBC via positive impact on inflammatory microenvironment by modulating COX-2 signal. As a prodrug, ketoplatin afforded 50.26-fold higher cytotoxicity than cisplatin against TNBC mesenchymal-stem cell-like MDA-MB-231 cells, partly attributing to its dramatic increase of cellular uptake and DNA damage. More importantly, EMT progress in MDA-MB-231 was markedly restrained by ketoplatin, resulting from the suppression of vimentin and N-cadherin mediated by down-regulated COX-2. Further in vivo investigation exhibited that ketoplatin effectively inhibited tumor growth and reduced systemic toxicity compared to cisplatin. Overall, ketoplatin possessed high antitumor activity and low toxicity against TNBC MDA-MB-231 in vitro and in vivo.


Asunto(s)
Antineoplásicos/administración & dosificación , Cisplatino/análogos & derivados , Cetoprofeno/administración & dosificación , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/metabolismo , Microambiente Tumoral/efectos de los fármacos , Células A549 , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/química , Antineoplásicos/química , Cisplatino/administración & dosificación , Cisplatino/química , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/fisiología , Femenino , Células HeLa , Humanos , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/metabolismo , Cetoprofeno/análogos & derivados , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Resultado del Tratamiento , Neoplasias de la Mama Triple Negativas/patología , Microambiente Tumoral/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
5.
Dalton Trans ; 50(1): 68-71, 2021 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-33320144

RESUMEN

Arsenoplatin-1 (AP-1) is an innovative dual-action anticancer agent that contains a platinum(ii) center coordinated to an arsenous acid moiety. We found that AP-1 spontaneously aggregates in aqueous solutions generating oligomeric species of increasing length. Afterward, we succeeded in solving the crystal structure of the adduct formed between the model protein lysozyme and an early AP-1 oligomer that turned out to be a trimer. Remarkably, this crystal structure traps an early stage of AP-1 aggregation offering detailed insight into the molecular process of the oligomer's growth.


Asunto(s)
Antineoplásicos/química , Trióxido de Arsénico/análogos & derivados , Arsenitos/química , Cisplatino/análogos & derivados , Complejos de Coordinación/química , Muramidasa/química , Platino (Metal)/química , Trióxido de Arsénico/química , Cisplatino/química , Estructura Molecular , Unión Proteica , Soluciones
6.
Carbohydr Polym ; 246: 116654, 2020 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-32747286

RESUMEN

Although some drug-based supramolecular systems have been constructed to overcome multidrug resistance and enhance the bioavailability of chemical drugs, strengthening the specific stimuli-responsive and active targeting ability of these systems is still a major challenge. In this paper, the synthesis and self-assembly behaviour of supramolecular self-assemblies with active targeting ß-cyclodextrin-modified hyaluronic acid (HA-CD) and drug-drug conjugates (curcumin-oxoplatin, Cur-Pt) as building moieties were carefully investigated. Notably, the curcumin was chosen not only as the chemical anti-cancer drug, but also acted as the guest molecule which could be included into CD cavity to form host-guest interaction-based supramolecular assemblies. The obtained self-assemblies exhibited pH- and esterase-responsive drug release behaviours. Furthermore, basic cell experiments were performed to prove their effective cellular toxicity based on A549 cells and PC3 cells with high expression of CD44 receptor but they showed no toxicity to normal LO-2 cells with low expression of CD44 receptor, which suggests their potential application in the targeted drug release field.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/análogos & derivados , Curcumina/farmacología , Preparaciones de Acción Retardada/síntesis química , Glicoconjugados/farmacología , Ácido Hialurónico/química , beta-Ciclodextrinas/química , Células A549 , Antineoplásicos/química , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cisplatino/química , Cisplatino/farmacología , Curcumina/química , Portadores de Fármacos , Liberación de Fármacos , Esterasas/química , Expresión Génica , Glicoconjugados/química , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Receptores de Hialuranos/genética , Receptores de Hialuranos/metabolismo , Concentración de Iones de Hidrógeno , Cinética , Especificidad de Órganos , Células PC-3
7.
Int J Mol Sci ; 21(13)2020 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-32635199

RESUMEN

Although Pt(II)-based drugs are widely used to treat cancer, very few molecules have been approved for routine use in chemotherapy due to their side-effects on healthy tissues. A new approach to reducing the toxicity of these drugs is generating a prodrug by increasing the oxidation state of the metallic center to Pt(IV), a less reactive form that is only activated once it enters a cell. We used theoretical tools to combine the parent Pt(IV) prodrug, oxoplatin, with the most recent FDA-approved anti-cancer drug set published by the National Institute of Health (NIH). The only prerequisite imposed for the latter was the presence of one carboxylic group in the structure, a chemical feature that ensures a link to the coordination sphere via a simple esterification procedure. Our calculations led to a series of bifunctional prodrugs ranked according to their relative stabilities and activation profiles. Of all the designed molecules, the combination of oxoplatin with aminolevulinic acid as the bioactive ligand emerged as the most promising strategy by which to design enhanced dual-potency oncology drugs.


Asunto(s)
Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Cisplatino/análogos & derivados , Profármacos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/farmacocinética , Cisplatino/administración & dosificación , Cisplatino/química , Cisplatino/farmacocinética , Simulación por Computador , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ligandos , Modelos Moleculares , Estructura Molecular , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Profármacos/química , Profármacos/farmacocinética , Termodinámica
8.
J Inorg Biochem ; 209: 111096, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32485478

RESUMEN

Quite surprisingly, cisplatin and cis-[PtI2(NH3)2] were found to manifest significant differences in their reactions with the model protein lysozyme. We decided to explore whether these differences recur when reacting these two Pt compounds with other proteins. Notably, ESI-MS measurements carried out on cytochrome c nicely confirmed the reaction pattern observed for lysozyme. This prompted us to exploit a computational DFT approach to disclose the molecular basis of such behavior. We analyzed comparatively the reactions of cis-[PtCl2(NH3)2] and cis-[PtI2(NH3)2] with appropriate molecular models (Ls) of the sidechains of relevant aminoacids. We found that when Pt(II) complexes are reacted with sulfur ligands both quickly lose their halide ligands and then the resulting cis-[Pt(L)2(NH3)2] species loses ammonia upon reaction with a ligand excess. In the case of imidazole, again cis-[PtCl2(NH3)2] and cis-[PtI2(NH3)2] quickly lose their halide ligands but the resulting cis-[Pt(L)2(NH3)2] species does not lose ammonia by reaction with excess imidazole. These results imply that the two platinum complexes manifest a significantly different behavior in their reaction with representative small molecules in agreement with what observed in the case of model proteins. It follows that the protein itself must play a crucial role in triggering the peculiar reactivity of cis-[PtI2(NH3)2] and in governing the nature of the formed protein adducts. The probable reasons for the observed behavior are critically commented and discussed.


Asunto(s)
Cisplatino/análogos & derivados , Cisplatino/química , Citocromos c/metabolismo , Aminoácidos/metabolismo , Amoníaco/metabolismo , Antineoplásicos/química , Antineoplásicos/metabolismo , Cisplatino/metabolismo , Imidazoles/metabolismo , Modelos Moleculares , Muramidasa/metabolismo , Compuestos Organoplatinos/química , Unión Proteica , Espectrometría de Masa por Ionización de Electrospray/métodos
9.
Molecules ; 25(3)2020 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-32033039

RESUMEN

A series of bile acid derived 1,2- and 1,3-diamines as well as their platinum(II) complexes were designed and synthesized in hope to get a highly cytotoxic compound by the combination of two bioactive moieties. All complexes obtained were subjected to cytotoxicity assays in vitro and some hybrid molecules showed an expected activity.


Asunto(s)
Ácidos y Sales Biliares/química , Cisplatino/análogos & derivados , Compuestos de Platino/síntesis química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Diseño de Fármacos , Células HeLa , Células Endoteliales de la Vena Umbilical Humana , Humanos , Concentración 50 Inhibidora , Células MCF-7 , Estructura Molecular , Compuestos de Platino/química , Compuestos de Platino/farmacología
10.
Dalton Trans ; 49(5): 1613-1619, 2020 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-31942585

RESUMEN

We herein designed two new PtIV prodrugs of oxoplatin (cis,cis,cis-[PtCl2(NH3)2(OH)2]), [PtIVCl2(NH3)2(O2C-FA)2] (Pt-2) and [PtIVCl2(NH3)2(O2C-RH)2] (Pt-3), by conjugating with ferulic acid (FA-COOH) and rhein (RH-COOH) which have well-known biological activities. Three other Pt(iv) complexes of [PtIVCl2(NH3)2(O2C-BA)2] (Pt-1), [PtIVCl2(NH3)2(O2C-CA)2] (Pt-4) and [PtIVCl2(NH3)2(O2C-TCA)2] (Pt-5) (where BA-COOH = benzoic acid, CA-COOH = crotonic acid and TCA-COOH = trans-cinnamic acid) were also prepared for the comparative study. Like most PtIV prodrug complexes, the cytotoxicity of Pt-3 containing the biologically active rhein (RH-COOH) ligand against lung carcinoma (A549 and A549/DDP) cells was higher than those of Pt-1, Pt-2, Pt-4, cisplatin and Pt-5. Moreover, the cytotoxicity of Pt-3 in HL-7702 normal cells was lower than those of PtIV derivatives bearing BA-COOH, FA-COOH, TCA-COOH and CA-COOH ligands. The highly efficacious Pt-2 and Pt-3 were found to accumulate strongly in the A549/DDP cells, with the prodrug Pt-3 showing highest levels of penetration into the mitochondria. The prodrug Pt-3 effectively entered the A549/DDP cells and caused mitochondrial damage, significantly greater than Pt-2. In addition, the prodrug Pt-3 exhibited higher antitumor efficacy (inhibition rates (IR) = 67.45%) than Pt-2 (28.12%) and cisplatin (33.05%) in the A549/DDP xenograft mouse model. Thus, the prodrug Pt-3 containing the rhein (RH-COOH) ligand is a promising candidate drug targeting the mitochondria.


Asunto(s)
Antraquinonas/farmacología , Antineoplásicos/farmacología , Cisplatino/análogos & derivados , Ácidos Cumáricos/farmacología , Compuestos Organoplatinos/farmacología , Profármacos/farmacología , Células A549 , Animales , Antraquinonas/química , Antineoplásicos/síntesis química , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cisplatino/química , Cisplatino/farmacología , Ácidos Cumáricos/química , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ligandos , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Compuestos Organoplatinos/síntesis química , Compuestos Organoplatinos/química , Profármacos/síntesis química , Profármacos/química , Relación Estructura-Actividad
11.
Exp Cell Res ; 388(1): 111813, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31904382

RESUMEN

Lysophosphatidic acid (LPA) signaling via LPA receptors (LPA1 to LPA6) regulates a variety of malignant properties of cancer cells. It is known that endothelial cells promote tumor progression and chemoresistance. The present study aimed to investigate the roles of LPA5 in cellular functions modulated by endothelial cells and anticancer drug in osteosarcoma cells. Human osteosarcoma MG-63 cells were maintained in endothelial F2 cell supernatants. After culturing for 3 months, MG63-F2 cells were established. LPAR5 expression level in MG63-F2 cells was significantly elevated, compared with MG-63 cells. The cell motile activity of MG63-F2 cells was markedly higher than that of MG-63 cells. To validate the effects of LPA5 on cell motile activity, LPA5 knockdown cells were generated from MG-63 cells. The cell motile activity of MG-63 cells was inhibited by LPA5 knockdown. The cell survival to cisplatin (CDDP) was reduced in MG-63 cells treated with LPA. In the presence of LPA, the cell survival rate was significantly lower in MG63-F2 cells than MG-63 cells, correlating with LPAR5 expression. LPA5 knockdown cells indicated the high cell survival rate to CDDP. Moreover, LPAR5 expression level was increased in the long-term CDDP treated MG63-C cells. The cell survival to CDDP of MG63-C cells was enhanced by LPA5 knockdown. These results suggest that cellular functions are regulated through LPA5-mediatd signaling induced by endothelial cells and CDDP in MG-63 cells.


Asunto(s)
Células Endoteliales/metabolismo , Osteosarcoma/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Antineoplásicos/farmacología , Muerte Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Cisplatino/análogos & derivados , Cisplatino/farmacología , Medios de Cultivo Condicionados/farmacología , Humanos , Lisofosfolípidos/metabolismo , Receptores del Ácido Lisofosfatídico/genética , Transducción de Señal , Células Tumorales Cultivadas
12.
Bioconjug Chem ; 30(6): 1658-1664, 2019 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-31070357

RESUMEN

Black phosphorus (BP) is a two-dimensional (2D) nanomaterial with high charge-carrier mobility, a tunable direct bandgap, and a unique in-plane anisotropic structure; however, the easiness of BP oxidation into P xO y species in ambient conditions largely limits its applications. In this study, modified cisplatin-Pt-NO3 [Pt(NH3)2(NO3)2] is used for surface coordination with BP nanosheets to generate Pt@BP, which maintains the surface morphology and properties of BP nanosheets for more than 24 h in ambient conditions. In addition, Pt@BP interacts with DNA both in vitro and in cell. Pt@BP shows a good cellular uptake rate and significantly increases the drug sensitivity of cisplatin-resistant cancer cell lines (A2780 and HepG2) compared with unmodified cisplatin. Our study is the first attempt to stabilize bare BP with cationic cisplatin species, and the generated Pt@BP could be used for potential synergistic photothermal/chemotherapy of cisplatin-resistant cancer.


Asunto(s)
Antineoplásicos/química , Cisplatino/análogos & derivados , Fósforo/química , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Cisplatino/síntesis química , Cisplatino/farmacología , Humanos , Nanoestructuras/química , Neoplasias/tratamiento farmacológico , Fósforo/farmacología
13.
J Med Chem ; 62(9): 4543-4554, 2019 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-31002510

RESUMEN

As FDA-approved chemotherapeutic agents, cisplatin, oxaliplatin, and 5-fluorouracil are widely used in clinic but limited by severe side-effects. To ameliorate their respective defects, a series of "dual-prodrug" by linking oxoplatin and 5-FU were designed and synthesized. The assembled compounds 10-17, named Fuplatin, exhibited much higher cytotoxicity against the tested cancer cells while lower cytotoxicity toward the human normal lung cells than free drugs or their combinations. Among them, 14 enhanced cellular accumulation with 62- and 825-fold amount of oxaliplatin and 8 at 9 h, respectively, significantly induced DNA damage and cell apoptosis, and inhibited migration and invasion in HCT-116 cells. Compound 14 arrested the cell cycle at S and G2 phases and up-regulated thymidylate synthase and p53, consistent with the results of the combination, suggesting 14 adopted a collaborative mode of 5-FU and oxaliplatin to kill cancer cells. In vivo, compound 14 showed high antitumor effect and no observable toxicity in NOD/SCID mice bearing HCT-116 tumors.


Asunto(s)
Antineoplásicos/uso terapéutico , Cisplatino/análogos & derivados , Fluorouracilo/análogos & derivados , Fluorouracilo/uso terapéutico , Profármacos/uso terapéutico , Animales , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Cisplatino/síntesis química , Cisplatino/farmacología , Cisplatino/uso terapéutico , Daño del ADN/efectos de los fármacos , Sinergismo Farmacológico , Fluorouracilo/farmacología , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Humanos , Ratones Endogámicos NOD , Ratones SCID , Profármacos/síntesis química , Profármacos/farmacología , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos , Timidilato Sintasa/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
14.
J Am Chem Soc ; 141(16): 6453-6457, 2019 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-30943017

RESUMEN

Arsenoplatins are adducts of two chemically important anticancer drugs, cisplatin and arsenic trioxide, that have a Pt(II) bond to an As(III) hydroxide center. Screens of the NCI-60 human tumor cell lines reveal that arsenoplatin-1 (AP-1), [Pt(µ-NHC(CH3)O)2ClAs(OH)2], the first representative of this novel class of anticancer agents, displays a superior activity profile relative to the parent drugs As2O3 or cisplatin in a majority of cancer cell lines tested. These activity profiles are important because the success of arsenic trioxide in blood cancers (such as APL) has not been seen in solid tumors due to the rapid clearance of arsenous acid from the body. To understand the biological chemistry of these compounds, we evaluated interactions of AP-1 with the two important classes of biomolecules-proteins and DNA. The first structural studies of AP-1 bound to model proteins reveal that platinum(II) binds the Nε of His in a manner that preserves the Pt-As bond. We find that AP-1 readily enters cells and binds to DNA with an intact Pt-As bond (Pt:As ratio of 1). At longer incubation times, however, the Pt:As ratio in DNA samples increases, suggesting that the Pt-As bond breaks and releases the As(OH)2 moiety. We conclude that arsenoplatin-1 has the potential to deliver both Pt and As species to a variety of hematological and solid cancers.


Asunto(s)
Antineoplásicos/farmacología , Trióxido de Arsénico/análogos & derivados , Cisplatino/análogos & derivados , Compuestos Organoplatinos/farmacología , Antineoplásicos/química , Trióxido de Arsénico/química , Trióxido de Arsénico/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cisplatino/química , Cisplatino/farmacología , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Estructura Molecular , Compuestos Organoplatinos/química , Relación Estructura-Actividad
15.
Metallomics ; 11(4): 833-844, 2019 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-30865741

RESUMEN

Interaction of platinum-based drugs with proteins containing sulphur amino acids is usually argued as one of the major reasons for the observed resistance to these drugs, mainly due to the deactivation of the native compounds by very efficient thiolation processes in the organism. In this work, we have investigated the detailed thermodynamics and kinetics of reaction between cisplatin cis-[PtCl2(NH3)2] and its major hydrolysed forms (monohydroxocisplatin cis-[PtCl(OH)(NH3)2] and monoaquacisplatin cis-[PtCl(H2O)(NH3)2]+) with various thiolates (methanethiolate, cysteine and glutathione) and methionine. We have used a demanding quantum chemistry approach at the MP2 and DFT levels of theory to determine the Gibbs free energies and the barrier of reactions of the most possible reaction paths. The substitution of the four ligands of the complexes studied here (Cl-, OH-, H2O and NH3) can either proceed by direct thiolations or bidentations. Our Raman spectroscopy measurements show that only two thiolations actually occur, although four are possible in principle. The reason could lie in the bidentation reactions eventually taking place after each thiolation, which is backed up by our computational results. The observed lability scale of the ligands under thiolate exposure was found to be in the following order H2O > Cl- ≈ NH3(trans) > NH3(cis) > OH-, the difference between ammine ligands being induced by a significant trans-labilization by thiolates. Finally, the S,N bidentation is shown to be preferred with respect to the S,O one.


Asunto(s)
Antineoplásicos/química , Cisplatino/química , Cisteína/química , Glutatión/química , Compuestos de Sulfhidrilo/química , Cisplatino/análogos & derivados , Hidrólisis , Modelos Moleculares , Teoría Cuántica , Termodinámica
16.
BMC Cancer ; 18(1): 1284, 2018 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-30577821

RESUMEN

BACKGROUND: Cisplatin has been widely used for the treatment of cancer and its antitumour activity is attributed to its capacity to form DNA adducts, predominantly at guanine residues, which impede cellular processes such as DNA replication and transcription. However, there are associated toxicity and drug resistance issues which plague its use. This has prompted the development and screening of a range of chemotherapeutic drug analogues towards improved efficacy. The biological properties of three novel platinum-based compounds consisting of varying cis-configured ligand groups, as well as a commercially supplied compound, were characterised in this study to determine their potential as anticancer agents. METHODS: The linear amplification reaction was employed, in conjunction with capillary electrophoresis, to quantify the sequence specificity of DNA adducts induced by these compounds using a DNA template containing telomeric repeat sequences. Additionally, the DNA interstrand cross-linking and unwinding efficiency of these compounds were assessed through the application of denaturing and native agarose gel electrophoresis techniques, respectively. Their cytotoxicity was determined in HeLa cells using a colorimetric cell viability assay. RESULTS: All three novel platinum-based compounds were found to induce DNA adduct formation at the tandem telomeric repeat sequences. The sequence specificity profile at these sites was characterised and these were distinct from that of cisplatin. Two of these compounds with the enantiomeric 1,2-diaminocyclopentane ligand (SS and RR-DACP) were found to induce a greater degree of DNA unwinding than cisplatin, but exhibited marginally lower DNA cross-linking efficiencies. Furthermore, the RR-isomer was more cytotoxic in HeLa cells than cisplatin. CONCLUSIONS: The biological characteristics of these compounds were assessed relative to cisplatin, and a variation in the sequence specificity and a greater capacity to induce DNA unwinding was observed. These compounds warrant further investigations towards developing more efficient chemotherapeutic drugs.


Asunto(s)
Aductos de ADN/efectos de los fármacos , ADN/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Compuestos Organoplatinos/química , Cisplatino/análogos & derivados , Cisplatino/química , Cisplatino/uso terapéutico , Reactivos de Enlaces Cruzados , ADN/química , Daño del ADN/efectos de los fármacos , Células HeLa , Humanos , Conformación de Ácido Nucleico/efectos de los fármacos , Compuestos Organoplatinos/uso terapéutico , Platino (Metal)/química , Platino (Metal)/uso terapéutico , Estereoisomerismo
17.
Biomater Sci ; 7(1): 262-271, 2018 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-30465673

RESUMEN

Cisplatin (CDDP) and arsenic trioxide (ATO), two representative inorganic anticancer drugs, have been successful in the treatment against several kinds of malignancies. However, combination therapy with these two drugs in clinical application suffers from poor pharmacokinetics, serious side effects, and drug resistance of the tumor. Herein, we report a carrier-free aquo-cisplatin arsenite multidrug nanocomposite loaded with cisplatin and arsenic trioxide prodrugs simultaneously. This nanocomposite achieves a high loading capacity and pH-dependent controlled release of the drugs. Because of these features, this nanocomposite shows better in vitro toxicity against various carcinoma cell lines than either the single drug or free drug combination, promotes the synergistic effect of cisplatin and arsenic trioxide, and significantly inhibits the growth of tumors in vivo. Furthermore, cisplatin and arsenic trioxide in this nanocomposite can realize a coordination of both enhanced DNA damage and DNA repair interference within cisplatin-resistant cells, which results in overcoming the drug resistance effectively. Gene expression profiles demonstrate the reduced expression of proto-oncogenes and DNA damage repair related genes MYC, MET, and MSH2, along with the increase of tumor suppressor genes PTEN, VHL, and FAS after the nanocomposite treatment. This type of multidrug nanocomposite offers an alternative and promising strategy for combination therapy and overcoming drug resistance.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Trióxido de Arsénico/análogos & derivados , Trióxido de Arsénico/farmacología , Cisplatino/análogos & derivados , Cisplatino/farmacología , Nanocompuestos/química , Animales , Antineoplásicos/síntesis química , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Trióxido de Arsénico/síntesis química , Trióxido de Arsénico/uso terapéutico , Arsenitos/síntesis química , Arsenitos/química , Arsenitos/farmacología , Línea Celular Tumoral , Cisplatino/síntesis química , Cisplatino/uso terapéutico , Daño del ADN/efectos de los fármacos , Resistencia a Antineoplásicos , Humanos , Masculino , Ratones Endogámicos BALB C , Nanocompuestos/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , Transcriptoma/efectos de los fármacos
18.
Gen Physiol Biophys ; 37(5): 515-525, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30307402

RESUMEN

The therapeutic use of cisplatin for the treatment of solid tumours is associated with organ toxicity. Amongst those, the cardiotoxicity is an occasional but very serious and severe side effect. To prevent or reduce these negative effects, many cisplatin analogues have been synthesized and evaluated in terms of being a less toxic and more effective agent. In present study, we examined the effects of cisplatin and its three analogues in the isolated rat heart to determine whether changes in the structure of the platinum complexes (changing of carrier ligands - ethylenediamine; 1,2-diaminocyclohexane; 2,2':6',2''-terpyridine) can influence their cardiotoxic effects. The results of our research indicate that the introduction of aromatic rings in the structure of the platinum complexes has a negative influence on the heart function. Conversely, the other two examined complexes had less negative effects on heart function compared to cisplatin. Our findings may be of interest for a possible synthetic strategy of introducing a carrier ligand that will exert a less cardiotoxic effect.


Asunto(s)
Cisplatino/análogos & derivados , Cisplatino/efectos adversos , Circulación Coronaria/efectos de los fármacos , Corazón/efectos de los fármacos , Corazón/fisiología , Perfusión , Animales , Relación Dosis-Respuesta a Droga , Hemodinámica/efectos de los fármacos , Masculino , Ratas , Ratas Wistar
19.
Biochem Pharmacol ; 156: 109-119, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30138622

RESUMEN

The efficacy of the anticancer drug cisplatin is restricted by tumor cell resistance and occurrence of severe side effects. One strategy to overcome these limitations is the development of new, improved platinum drugs. Previous investigations showed that platinum(IV)-nitroxyl complexes are able to circumvent cisplatin resistance in bladder cancer cells. In the present study the mode of action of the platinum(IV)-nitroxyl complex PN149 was investigated in the bladder cancer cell line RT112 and the renal cell carcinoma cell line A498 on the molecular and cellular level. Gene expression analysis showed that PN149 induced genes related to DNA damage response (RRM2B, GADD45A), cell cycle regulation (CDKN1A, PLK3, PPM1D) as well as those coding for the pro-apoptotic factors PUMA and Noxa. These findings on the transcriptional level were confirmed on the functional level revealing that PN149 treatment increased levels of p53 and resulted in cell cycle arrest and drug-induced cytotoxicity via induction of apoptosis. Regarding the expression of oxidative-stress sensitive genes, PN149 induced FTH1, GCLC, HMOX1 and TXNRD1 but relevant effects were restricted to RT112 cells treated with 50 µM. The pro-inflammatory IL-8 was induced by PN149 in RT112 but not A498 cells indicating a cell-type specific activation. Taken together, PN149 possessed promising activity in different tumor cell lines rendering it an interesting alternative to cisplatin in chemotherapy.


Asunto(s)
Cisplatino/análogos & derivados , Cisplatino/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma de Células Renales , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cisplatino/química , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Estructura Molecular , Neoplasias de la Vejiga Urinaria
20.
Molecules ; 23(6)2018 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-29890688

RESUMEN

Three new crystal structures containing [PtCl6]2−, pyridinium and benzimidazole groups have been prepared: [PtCl6]·(H-bzm)2·2(H2O) (1), [PtCl6]·(H-bipy)2·2(H2O) (2), [PtCl6]·(H-dimethyl-bipy)2·2(H2O) (3) [H-bzm: benzimidazole cation, H-bipy: 2,2'-bipyridine cation, H-dimethyl-bipy: 4,4'-bimethyl-2,2'-bipyridine cation]. All compounds have been fully characterized by elemental analyses, single-crystal X-ray analyses, IR spectra, TG analyses, and fluorescence studies. Single-crystal X-ray diffraction analysis suggests that the primary synthon contains ⁺N⁻H···Cl−, including ionic bonding and hydrogen bonding interactions. The dimensions are enhanced further by secondary O⁻H ∙∙Cl and N⁻H ∙∙O hydrogen bonding interactions between donor and acceptor atoms located at the periphery of these synthons. Moreover, coulombic attractions between the ions play an important role in reinforcing the structures of these complexes. In addition, antitumor activity against human lung adenocarcinoma cell line (A549) and human nasopharyngeal carcinoma cell line (CNE-2) was performed. These complexes all showed inhibition to the two cell lines, while complex 3 exhibited higher efficiency than complexes 1⁻2.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Cisplatino/análogos & derivados , Compuestos Heterocíclicos/química , Compuestos Inorgánicos/química , Nitrógeno/química , Compuestos Orgánicos/química , Antineoplásicos/síntesis química , Cationes , Cisplatino/síntesis química , Cisplatino/química , Cisplatino/farmacología , Cristalografía por Rayos X , Enlace de Hidrógeno , Estructura Molecular , Espectrofotometría Ultravioleta , Termogravimetría
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...