Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Adv Sci (Weinh) ; 9(13): e2105506, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35246961

RESUMEN

Membrane-lytic peptides offer broad synthetic flexibilities and design potential to the arsenal of anticancer therapeutics, which can be limited by cytotoxicity to noncancerous cells and induction of drug resistance via stress-induced mutagenesis. Despite continued research efforts on membrane-perforating peptides for antimicrobial applications, success in anticancer peptide therapeutics remains elusive given the muted distinction between cancerous and normal cell membranes and the challenge of peptide degradation and neutralization upon intravenous delivery. Using triple-negative breast cancer as a model, the authors report the development of a new class of anticancer peptides. Through function-conserving mutations, the authors achieved cancer cell selective membrane perforation, with leads exhibiting a 200-fold selectivity over non-cancerogenic cells and superior cytotoxicity over doxorubicin against breast cancer tumorspheres. Upon continuous exposure to the anticancer peptides at growth-arresting concentrations, cancer cells do not exhibit resistance phenotype, frequently observed under chemotherapeutic treatment. The authors further demonstrate efficient encapsulation of the anticancer peptides in 20 nm polymeric nanocarriers, which possess high tolerability and lead to effective tumor growth inhibition in a mouse model of MDA-MB-231 triple-negative breast cancer. This work demonstrates a multidisciplinary approach for enabling translationally relevant membrane-lytic peptides in oncology, opening up a vast chemical repertoire to the arms race against cancer.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama Triple Negativas , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Humanos , Ratones , Péptidos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/metabolismo
2.
Chembiochem ; 21(19): 2854-2860, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32415808

RESUMEN

We report the anti-osteosarcoma and anti-osteosarcoma stem cell (OSC) properties of a nickel(II) complex, 1. Complex 1 displays similar potency towards bulk osteosarcoma cells and OSCs, in the micromolar range. Notably, 1 displays similar or better OSC potency than the clinically approved platinum(II) anticancer drugs cisplatin and carboplatin in two- and three-dimensional osteosarcoma cell cultures. Mechanistic studies revealed that 1 induces osteosarcoma cell death by necroptosis, an ordered form of necrosis. The nickel(II) complex, 1 triggers necrosome-dependent mitrochondrial membrane depolarisation and propidium iodide uptake. Interestingly, 1 does not evoke necroptosis by elevating intracellular reactive oxygen species (ROS) or hyperactivation of poly ADP ribose polymerase (PARP-1). ROS elevation and PARP-1 activity are traits that have been observed for established necroptosis inducers such as shikonin, TRAIL and glutamate. Thus the necroptosis pathway evoked by 1 is distinct. To the best of our knowledge, this is the first report into the anti-osteosarcoma and anti-OSC properties of a nickel complex.


Asunto(s)
Antineoplásicos/farmacología , Complejos de Coordinación/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Níquel/farmacología , Osteosarcoma/tratamiento farmacológico , Fenantrolinas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Supervivencia Celular/efectos de los fármacos , Complejos de Coordinación/síntesis química , Complejos de Coordinación/química , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Estructura Molecular , Necroptosis/efectos de los fármacos , Níquel/química , Osteosarcoma/metabolismo , Osteosarcoma/patología , Fenantrolinas/química , Especies Reactivas de Oxígeno/metabolismo , Relación Estructura-Actividad , Células Tumorales Cultivadas
4.
Chem Sci ; 10(33): 7792-7800, 2019 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-31588328

RESUMEN

Intracellular redox modulation offers a viable approach to effectively remove cancer stem cells (CSCs), a subpopulation of tumour cells thought to be responsible for cancer recurrence and metastasis. Here we report the breast CSC potency of reactive oxygen species (ROS)-generating manganese(ii)- and copper(ii)-4,7-diphenyl-1,10-phenanthroline complexes bearing diclofenac, a nonsteriodial anti-inflammatory drug (NSAID), 1 and 3. Notably, the manganese(ii) complex, 1, exhibits 9-fold, 31-fold, and 40-fold greater potency towards breast CSCs than 3, salinomycin (an established breast CSC-potent agent), and cisplatin (a clinically approved anticancer drug) respectively. Encouragingly, 1 displays 61-fold higher potency toward breast CSCs than normal skin fibroblast cells. Clinically relevant epithelial spheroid studies show that 1 is able to selectively inhibit breast CSC-enriched HMLER-shEcad mammosphere formation and viability (one order of magnitude) over non-tumorigenic breast MCF10A spheroids. Mechanistic studies show that 1 prompts breast CSC death by generating intracellular ROS and inhibiting cyclooxygenase-2 (COX-2) activity. The manganese(ii) complex, 1, induces a greater degree of intracellular ROS in CSCs than the corresponding copper(ii) complex, 3, highlighting the ROS-generating superiority of manganese(ii)- over copper(ii)-phenanthroline complexes. Encapsulation of 1 by biodegradable methoxy poly(ethylene glycol)-b-poly(d,l-lactic-co-glycolic) acid (PEG-PLGA) copolymers at the appropriate feed (5%, 1 NP5 ) enhances breast CSC uptake and greatly reduces overall toxicity. The nanoparticle formulation 1 NP5 indiscriminately kills breast CSCs and bulk breast cancer cells, and evokes a similar cellular response to the payload, 1. To the best of our knowledge, this is the first study to investigate the anti-CSC properties of managense complexes and to demonstrate that polymeric nanoparticles can be used to effectively deliver managense complexes into CSCs.

5.
Angew Chem Int Ed Engl ; 58(35): 12059-12064, 2019 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-31209956

RESUMEN

The preparation of multinuclear metal complexes offers a route to novel anticancer agents and delivery systems. The potency of a novel triangular multinuclear complex containing three platinum atoms, Pt-3, towards breast cancer stem cells (CSCs) is reported. The trinuclear platinum(II) complex, Pt-3 exhibits selective toxicity towards breast CSCs over bulk breast cancer cells and non-tumorigenic breast cells. Remarkably, Pt-3 inhibits the formation, size, and viability of mammospheres to a better extent than salinomycin, an established CSC-potent agent, and cisplatin and carboplatin, clinically used platinum drugs. Mechanism of action studies show that Pt-3 effectively enters breast CSCs, penetrates the nucleus, induces genomic DNA damage, and prompts caspase-dependent apoptosis. To the best of our knowledge, Pt-3 is the first multinuclear platinum complex to selectively kill breast CSCs over other breast cell types.


Asunto(s)
Apoptosis/efectos de los fármacos , Complejos de Coordinación/farmacología , Platino (Metal)/química , Antineoplásicos/química , Antineoplásicos/farmacología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Cadherinas/antagonistas & inhibidores , Cadherinas/genética , Cadherinas/metabolismo , Carboplatino/farmacología , Caspasas/metabolismo , Línea Celular Tumoral , Cisplatino/farmacología , Complejos de Coordinación/química , Cristalografía por Rayos X , Femenino , Humanos , Conformación Molecular , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Piranos/farmacología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo
6.
Molecules ; 24(9)2019 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-31035718

RESUMEN

Copper(II) complexes bearing nonsteroidal anti-inflammatory drugs (NSAIDs) are known to potently kill cancer stem cells (CSCs), a subpopulation of tumour cells with high metastatic and relapse fidelity. One of the major disadvantages associated to these copper(II) complexes is their instability in the presence of strong cellular reductants (such as ascorbic acid). Here we present a biologically stable copper(II)-NSAID complex containing a bathocuproinedisulfonic acid disodium ligand and two indomethacin moieties, Cu(bathocuproinedisulfonic acid disodium)(indomethacin)2, 2. The copper(II) complex, 2 kills bulk breast cancer cells and breast CSC equally (in the sub-micromolar range) and displays very low toxicity against non-tumorigenic breast and kidney cells (IC50 value > 100 µM). Three-dimensional cell culture studies show that 2 can significantly reduce the number and size of breast CSC mammospheres formed (from single suspensions) to a similar level as salinomycin (an established anti-breast CSC agent). The copper(II) complex, 2 is taken up reasonably by breast CSCs and localises largely in the cytoplasm (>90%). Cytotoxicity studies in the presence of specific inhibitors suggest that 2 induces CSC death via a reactive oxygen species (ROS) and cyclooxygenase isoenzyme-2 (COX-2) dependent apoptosis pathway.


Asunto(s)
Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/farmacología , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Cobre/química , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Complejos de Coordinación/síntesis química , Ciclooxigenasa 2/metabolismo , Humanos , Concentración 50 Inhibidora , Estructura Molecular , Especies Reactivas de Oxígeno/metabolismo , Análisis Espectral
7.
Dalton Trans ; 48(18): 5892-5896, 2019 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-30632590

RESUMEN

Copper(ii) coordination complexes, 1 and 2, containing nonsteroidal anti-inflammatory drugs (NSAIDs) potently kill breast cancer stem cells (CSCs) and bulk breast cancer cells. Although detailed biological studies have been conducted to shed light on their mechanism of cytotoxicity, little is known about their molecular level mechanism of action. This biophysical study, aided by the preparation of a fluorophore-containing analogue, 3, reveals that the complexes operate by undergoing reduction to a copper(i) form and releasing the associated NSAIDs.


Asunto(s)
Antineoplásicos/química , Fenómenos Biofísicos/efectos de los fármacos , Complejos de Coordinación/química , Cobre/química , Células Madre Neoplásicas/efectos de los fármacos , Antineoplásicos/farmacología , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/tratamiento farmacológico , Supervivencia Celular/efectos de los fármacos , Complejos de Coordinación/farmacología , Femenino , Colorantes Fluorescentes/química , Humanos , Indometacina/química , Naproxeno/química , Imagen Óptica/métodos , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo
8.
Dalton Trans ; 47(39): 13761-13765, 2018 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-30225483

RESUMEN

We report a novel series of cobalt(iii)-polypridyl complexes, 4-6, that can selectively release diflunisal, a nonsteroidal anti-inflammatory drug, under reducing conditions. Remarkably, the 1,10-phenanthroline-bearing complex 5 displays selective potency towards hard-to-kill cancer stem cells (CSCs) (IC50 = 2.1 ± 0.1 µM) over bulk cancer (IC50 = 3.9 ± 0.2 µM) and normal cells (IC50 = 21.2 ± 1.3 µM). This complex induces CSC apoptosis by DNA damage and cyclooxygenase-2 inhibition.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Cobalto/química , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Diflunisal/química , Células Madre Neoplásicas/efectos de los fármacos , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Humanos , Concentración 50 Inhibidora , Células Madre Neoplásicas/patología
9.
Molecules ; 23(9)2018 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-30181492

RESUMEN

Cancer stem cells (CSCs) are thought of as a clinically pertinent subpopulation of tumors, partly responsible for cancer relapse and metastasis. Research programs aimed at discovering anti-CSC agents have largely focused on biologics and purely organic molecules. Recently, we showed that a family of redox-active copper(II) complexes with phenanthroline-based ligands and nonsteroidal anti-inflammatory drugs (NSAIDs) such as indomethacin, are capable of potently and selectively killing breast CSCs. Herein we present analogous redox-inactive, zinc(II)-phenanthroline-indomethacin complexes with the ability to kill breast CSCs and bulk breast cancer cells with equal potency (in the submicro- or micromolar range). A single dose of the zinc(II) complexes could theoretically be administered to eliminate whole tumor populations. Excitingly, some of the zinc(II) complexes decrease the growth and viability of mammospheres to a comparable or higher degree than salinomycin, a compound known to effectively kill breast CSCs. As far as we are aware this is the first report to examine the anti-breast CSC activity of zinc(II)-containing compounds.


Asunto(s)
Neoplasias de la Mama/patología , Indometacina/farmacología , Células Madre Neoplásicas/patología , Piridinas/farmacología , Zinc/farmacología , Supervivencia Celular/efectos de los fármacos , Femenino , Células HEK293 , Humanos , Concentración 50 Inhibidora , Células Madre Neoplásicas/efectos de los fármacos , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/patología , Células Tumorales Cultivadas
10.
Chemistry ; 24(57): 15205-15210, 2018 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-30052298

RESUMEN

The cancer stem cell (CSC) toxicity and mechanism of action of a series of iridium(III) complexes bearing polypridyl and charged 1-methyl-2-(2-pyridyl)pyridinium ligands, 1-4 is reported. The most effective complex (containing 1,10-phenanthroline), 3, kills CSCs and bulk cancer cells with equal potency (in the micromolar range), indicating that it could potentially remove heterogenous tumour populations with a single dose. Encouragingly, 3 also inhibits mammopshere formation to a similar extent as salinomycin, a well-established anti-CSC agent. This complex induces CSC apoptosis by mitochondrial membrane depolarization, inhibition of mitochondrial metabolism, and intracellular reactive oxygen species (ROS) generation. To the best of our knowledge, this is the first study to investigate the anti-CSC properties of iridium complexes.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Iridio/química , Iridio/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Mitocondrias/efectos de los fármacos , Modelos Moleculares , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Fenantrolinas/química , Fenantrolinas/farmacología , Especies Reactivas de Oxígeno/metabolismo
11.
Dalton Trans ; 47(16): 5755-5763, 2018 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-29634060

RESUMEN

We report the synthesis and characterisation of mono- and di-nuclear cobalt(ii) complexes (1-3) containing L1, a polypyridyl ligand with pyrazole moieties. DNA binding studies suggest that the mono-nuclear complex, 1, binds to DNA via the grooves prior to inducing oxidative DNA cleavage whereas the larger di-nuclear complexes, 2 and 3, bind to DNA via the grooves and through intercalation prior to inducing oxidative DNA cleavage. The cobalt(ii) complexes display micromolar potency towards U2OS (bone osteosarcoma), HepG2 (liver hepatocellular carcinoma), and GM05757 (normal human fibroblast) cells, comparable to clinically used platinum agents, cisplatin and carboplatin. The cellular mechanism of action studies show that the most effective cobalt(ii) complex, 2, enters U2OS cells, penetrates the nucleus, induces genomic DNA damage, and triggers caspase-dependent apoptosis in a p53-independent manner. This study highlights the potential of di-nuclear cobalt(ii) complexes as artificial oxidative metallonucleases and tangible cancer cell-potent agents.

12.
Angew Chem Int Ed Engl ; 57(1): 287-291, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29144008

RESUMEN

The breast cancer stem cell (CSC) and bulk breast cancer cell potency of a series of metallopeptides containing dichloro(1,10-phenanthroline)copper(II) and various organelle-targeting peptide sequences is reported. The mitochondria-targeting metallopeptide 1 exploits the higher mitochondrial load in breast CSCs over the corresponding non-CSCs and the vulnerability of breast CSCs to mitochondrial damage to potently and selectively kill breast CSCs. Strikingly, 1 reduces the formation and size of mammospheres to a greater extent than salinomycin, an established CSC-potent agent. Mechanistic studies show that 1 enters CSC mitochondria, induces mitochondrial dysfunction, generates reactive oxygen species (ROS), activates JNK and p38 pathways, and prompts apoptosis. To the best of our knowledge, 1 is the first metallopeptide to selectivity kill breast CSCs in vitro.


Asunto(s)
Neoplasias de la Mama/patología , Complejos de Coordinación/farmacología , Metaloproteínas/farmacología , Mitocondrias/efectos de los fármacos , Células Madre Neoplásicas/patología , Péptidos/farmacología , Fenantrolinas/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , Femenino , Humanos , Concentración 50 Inhibidora , MAP Quinasa Quinasa 4/metabolismo , Metaloproteínas/química , Piranos/farmacología , Especies Reactivas de Oxígeno/metabolismo , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
Chemistry ; 23(47): 11366-11374, 2017 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-28658520

RESUMEN

Four copper(II) complexes, 1-4 containing regioisomeric vanillin Schiff base derivatives and the nonsteroidal anti-inflammatory drug (NSAID), naproxen, were synthesised and characterised. All complexes effectively cleave DNA in cell-free systems, with 4 displaying the highest nuclease activity. DNA binding studies suggest that 4 binds to DNA via the grooves prior to inducing oxidative DNA cleavage. Three of the complexes (1, 3, and 4) indiscriminately kill cancer stem cell (CSC)-enriched cells (HMLER-shEcad) and bulk cancer cells (HMLER) at micromolar concentrations. The most effective complex, 4 also reduced the formation and size of mammospheres to a similar extent as salinomycin, a well-established CSC-potent agent. Mechanistic studies show that 4 is readily taken up by CSCs, elevates intracellular reactive oxygen species (ROS) levels, causes DNA damage, and induces caspase-dependent apoptosis. Furthermore, 4 inhibits cyclooxygenase-2 (COX-2) expression and causes COX-2-dependent CSC death. The advantage of 4 over bulk cancer cell- or CSC-selective agents is that it has the potential to remove whole tumor populations (bulk cancer cells and CSCs) with a single dose.


Asunto(s)
Benzaldehídos/química , Complejos de Coordinación/química , Cobre/química , Naproxeno/química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Complejos de Coordinación/síntesis química , Complejos de Coordinación/toxicidad , Ciclooxigenasa 2/metabolismo , ADN/química , ADN/metabolismo , División del ADN/efectos de los fármacos , Humanos , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Bases de Schiff/química , Espectrofotometría
14.
Chemistry ; 23(40): 9674-9682, 2017 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-28556445

RESUMEN

The cytotoxic properties of a series of nickel(II)-dithiocarbamate phenanthroline complexes is reported. The complexes 1-6 kill bulk cancer cells and cancer stem cells (CSCs) with micromolar potency. Two of the complexes, 2 and 6, kill twice as many breast cancer stem cell (CSC)-enriched HMLER-shEcad cells as compared to breast CSC-depleted HMLER cells. Complex 2 inhibits mammosphere formation to a similar extent as salinomycin (a CSC-specific toxin). Detailed mechanistic studies suggest that 2 induces CSC death by necroptosis, a programmed form of necrosis. Specifically, 2 triggers MLKL phosphorylation, oligomerization, and translocation to the cell membrane. Additionally, 2 induces necrosome-mediated propidium iodide (PI) uptake and mitochondrial membrane depolarisation, as well as morphological changes consistent with necroptotosis. Strikingly, 2 does not evoke necroptosis by intracellular reactive oxygen species (ROS) production or poly(ADP) ribose polymerase (PARP-1) activation.


Asunto(s)
Antineoplásicos/síntesis química , Apoptosis/efectos de los fármacos , Complejos de Coordinación/síntesis química , Células Madre Neoplásicas/efectos de los fármacos , Níquel/química , Fenantrolinas/síntesis química , Tiocarbamatos/síntesis química , Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular , Complejos de Coordinación/farmacología , Humanos , Necrosis , Células Madre Neoplásicas/patología , Fenantrolinas/farmacología , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Tiocarbamatos/farmacología , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores
15.
Dalton Trans ; 45(44): 17867-17873, 2016 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-27774561

RESUMEN

We report the cancer stem cell (CSC) potency of a novel series of copper(ii)-phenanthroline complexes bearing nonsteriodial anti-inflammatory drugs: naproxen, tolfenamic acid, and indomethacin (2a-3c). Two of the complexes, 2a and 3c, kill breast CSC-enriched HMLER-shEcad cells (grown in both monolayer and three-dimensional cell cultures) to a significantly better extent than salinomycin, a well-established CSC toxin. The most potent complex in the series, 3c induces its cytotoxic effect by generating intracellular reactive oxygen species (ROS) and inhibiting cyclooxgenase-2 (COX-2) activity. Encapsulation of 3c using biodegradable methoxy poly(ethylene glycol)-b-poly(d,l-lactic-co-glycolic) acid (PEG-PLGA) copolymers at the appropriate feed (5%, 3c NP5) enhances breast CSC uptake and reduces overall toxicity. The nanoparticle formulation, 3c NP5 selectively kills breast CSCs over bulk breast cancer cells, and evokes a similar cellular response to the payload, 3c. To the best of our knowledge, this is the first study to demonstrate that polymeric nanoparticles can be used to effectively deliver CSC-potent metal complexes into CSCs.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Complejos de Coordinación/farmacología , Cobre/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/química , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Neoplasias de la Mama/patología , Línea Celular , Línea Celular Tumoral , Complejos de Coordinación/administración & dosificación , Complejos de Coordinación/química , Cobre/administración & dosificación , Cobre/química , Portadores de Fármacos/química , Femenino , Humanos , Nanopartículas/química , Células Madre Neoplásicas/patología , Poliésteres/química , Polietilenglicoles/química
16.
Chembiochem ; 17(18): 1713-8, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27377813

RESUMEN

We report the potency against cancer stem cells (CSCs) of a new cobalt(III)-cyclam complex (1) that bears the nonsteroidal anti-inflammatory drug, naproxen. The complex displays selective potency for breast CSC-enriched HMLER-shEcad cells over breast CSC-depleted HMLER cells. Additionally, it inhibited the formation of three-dimensional tumour-like mammospheres, and reduced their viability to a greater extent than clinically used breast cancer drugs (vinorelbine, cisplatin and paclitaxel). The anti-mammosphere potency of 1 was enhanced under hypoxia-mimicking conditions. Detailed mechanistic studies revealed that DNA damage and cyclooxygenase-2 (COX-2) inhibition contribute to the cytotoxic mechanism of 1. To the best of our knowledge, 1 is the first cobalt-containing compound to show selective potency for CSCs over bulk cancer cells.


Asunto(s)
Antineoplásicos/farmacología , Cobalto/farmacología , Inhibidores de la Ciclooxigenasa 2/farmacología , Naproxeno/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Compuestos Organometálicos/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cobalto/química , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa 2/síntesis química , Inhibidores de la Ciclooxigenasa 2/química , Daño del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Conformación Molecular , Naproxeno/química , Compuestos Organometálicos/síntesis química , Compuestos Organometálicos/química , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...