Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Chem Commun (Camb) ; 60(48): 6150-6153, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38804255

RESUMO

Iron-binding strategies in anticancer drug design target the key role of iron in cancer growth. The incorporation of a quinoline moiety in the design of tetrazolium-based prochelators facilitates their intracellular reduction/activation to iron-binding formazans. The new prochelators are antiproliferative at submicromolar levels, induce apoptosis and cell cycle arrest, and impact iron signaling in cancer cells.


Assuntos
Antineoplásicos , Apoptose , Proliferação de Células , Ferro , Quinolinas , Humanos , Quinolinas/química , Quinolinas/farmacologia , Proliferação de Células/efeitos dos fármacos , Antineoplásicos/farmacologia , Antineoplásicos/química , Antineoplásicos/síntese química , Ferro/química , Ferro/metabolismo , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Estrutura Molecular , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos
2.
J Am Chem Soc ; 145(28): 15197-15206, 2023 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-37410992

RESUMO

Cancer cells generally present a higher demand for iron, which plays crucial roles in tumor progression and metastasis. This iron addiction provides opportunities to develop broad spectrum anticancer drugs that target iron metabolism. In this context, prochelation approaches are investigated to release metal-binding compounds under specific conditions, thereby limiting off-target toxicity. Here, we demonstrate a prochelation strategy inspired by the bioreduction of tetrazolium cations widely employed to assess the viability of mammalian cells. We designed a series of tetrazolium-based compounds for the intracellular release of metal-binding formazan ligands. The combination of reduction potentials appropriate for intracellular reduction and an N-pyridyl donor on the formazan scaffold led to two effective prochelators. The reduced formazans bind as tridentate ligands and stabilize low-spin Fe(II) centers in complexes of 2:1 ligand-to-metal stoichiometry. The tetrazolium salts are stable in blood serum for over 24 h, and antiproliferative activities at micromolar levels were recorded in a panel of cancer cell lines. Additional assays confirmed the intracellular activation of the prochelators and their ability to affect cell cycle progression, induce apoptotic death, and interfere with iron availability. Demonstrating the role of iron in their intracellular effects, the prochelators impacted the expression levels of key iron regulators (i.e., transferrin receptor 1 and ferritin), and iron supplementation mitigated their cytotoxicity. Overall, this work introduces the tetrazolium core as a platform to build prochelators that can be tuned for activation in the reducing environment of cancer cells and produce antiproliferative formazan chelators that interfere with cellular iron homeostasis.


Assuntos
Quelantes de Ferro , Ferro , Animais , Formazans , Quelantes de Ferro/química , Quelantes de Ferro/farmacologia , Ligantes , Ferro/química , Sais de Tetrazólio , Mamíferos/metabolismo
3.
Inorg Chem ; 61(49): 19974-19982, 2022 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-36455205

RESUMO

Tridentate thiosemicarbazones, among several families of iron chelators, have shown promising results in anticancer drug discovery because they target the increased need for iron that characterizes malignant cells. Prochelation strategies, in which the chelator is released under specific conditions, have the potential to avoid off-target metal binding (for instance, in the bloodstream) and minimize unwanted side effects. We report a prochelation approach that employs arylsulfonate esters to mask the phenolate donor of salicylaldehyde-based chelators. The new prochelators liberate a tridentate thiosemicarbazone intracellularly upon reaction with abundant nucleophile glutathione (GSH). A 5-bromo-substituted salicylaldehyde thiosemicarbazone (STC4) was selected for the chelator unit because of its antiproliferative activity at low micromolar levels in a panel of six cancer cell lines. The arylsulfonate prochelators were assessed in vitro with respect to their stability, ability to abolish metal binding, and reactivity in the presence of GSH. Cell-based assays indicated that the arylsulfonate-masked prochelators present higher antiproliferative activities relative to the parent compound after 24 h. The activation and release of the chelator intracellularly were corroborated by assays of cytosolic iron binding and iron supplementation effects as well as cell cycle analysis. This study introduces the 1,3,4-thiadiazole sulfonate moiety to mask the phenolate donor of an iron chelator and impart good solubility and stability to prochelator constructs. The reactivity of these systems can be tuned to release the chelator at high glutathione levels, as encountered in several cancer phenotypes.


Assuntos
Antineoplásicos , Tiossemicarbazonas , Ferro/química , Compostos de Sulfidrila/farmacologia , Quelantes de Ferro/farmacologia , Quelantes de Ferro/química , Tiossemicarbazonas/química , Glutationa/metabolismo , Linhagem Celular , Antineoplásicos/farmacologia , Antineoplásicos/química
4.
ACS Med Chem Lett ; 13(9): 1452-1458, 2022 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-36105345

RESUMO

Glycoconjugation strategies in anticancer drug discovery exploit the high expression of glucose transporters in malignant cells to achieve preferential uptake and hence attractive pharmacological characteristics of increased therapeutic windows and decreased unwanted toxicity. Here we present the design of glycoconjugated prochelators of aroylhydrazone AH1, an antiproliferative scavenger that targets the increased iron demand of rapidly proliferating malignant cells. The constructs feature a monosaccharide (d-glucose, d-glucosamine, or glycolytic inhibitor 2-deoxy-d-glucose) connected at the C2 or C6 position via a short linker, which masks the chelator through a disulfide bond susceptible to intracellular reduction. Cellular assays showed that the glycoconjugates rely on the GLUT1 transporter for uptake, lead to intracellular iron deprivation, and present antiproliferative activity. Ectopic overexpression of GLUT1 in malignant and normal cells increased the uptake and toxicity of the glycoconjugated prochelators, demonstrating that these compounds are well suited for targeting cells overexpressing glucose transporters and therefore for selective iron sequestration in malignant cells.

5.
ChemMedChem ; 16(18): 2764-2768, 2021 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-33974730

RESUMO

The central role of iron in tumor progression and metastasis motivates the development of iron-binding approaches in cancer chemotherapy. Disulfide-based prochelators are reductively activated upon cellular uptake to liberate thiol chelators responsible for iron sequestration. Herein, a trimethyl thiosemicarbazone moiety and the imidazole-2-thione heterocycle are incorporated in this prochelator design. Iron binding of the corresponding tridentate chelators leads to the stabilization of a low-spin ferric center in 2 : 1 ligand-to-metal complexes. Native mass spectrometry experiments show that the prochelators form stable disulfide conjugates with bovine serum albumin, thus affording novel bioconjugate prochelator systems. Antiproliferative activities at sub-micromolar levels are recorded in a panel of breast, ovarian and colorectal cancer cells, along with significantly lower activity in normal fibroblasts.


Assuntos
Antineoplásicos/farmacologia , Imidazóis/farmacologia , Ferro/farmacologia , Tionas/farmacologia , Tiossemicarbazonas/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Imidazóis/química , Ferro/química , Ligantes , Estrutura Molecular , Relação Estrutura-Atividade , Tionas/química , Tiossemicarbazonas/química
6.
Inorg Chem ; 59(16): 11377-11384, 2020 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-32799490

RESUMO

Tridentate aroyl hydrazones are effective metal chelators in biological settings, and their activity has been investigated extensively for medicinal applications in metal overload, cancer, and neurodegenerative diseases. The aroyl hydrazone motif is found in the recently reported prochelator (AH1-S)2, which has shown antiproliferative proapoptotic activity in mammalian cancer cell lines. Intracellular reduction of this disulfide prochelator leads to the formation of mercaptobenzaldehyde benzoylhydrazone chelator AH1 and to iron sequestration, which in turn impacts cell growth. Herein, we investigate the iron coordination chemistry of AH1 to determine the structural and spectroscopic properties of the iron complexes in the solid state and in solution. A neutral iron(III) complex of 2:1 ligand-to-metal stoichiometry was isolated and characterized fully to reveal two different binding modes for the tridentate AH1 ligand. Specifically, one ligand binds in the monoanionic keto form, whereas the other ligand coordinates as a dianionic enolate. Continuous-wave electron paramagnetic resonance experiments in frozen solutions indicated that this neutral complex is one of three low-spin iron(III) complexes observed depending on the pH of the solution. Electron spin echo envelope modulation (ESEEM) experiments allowed assignment of the three species to different protonation states of the coordinated ligands. Our ESEEM analysis provides a method to distinguish the coordination of aroyl hydrazones in the keto and enolate forms, which influences both the ligand field and overall charge of the complex. As such, this type of analysis could provide valuable information in a variety of studies of iron complexes of aroyl hydrazones, ranging from the investigation of spin-crossover behavior to tracking of their distribution in biological samples.


Assuntos
Proliferação de Células/efeitos dos fármacos , Espectroscopia de Ressonância de Spin Eletrônica/métodos , Hidrazonas/química , Hidrazonas/farmacologia , Ferro/química , Linhagem Celular Tumoral , Cristalografia por Raios X , Feminino , Humanos , Estrutura Molecular , Prótons
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA