Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Inorg Chem ; 63(15): 6822-6835, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38560761

RESUMO

Boron-dipyrromethene (BODIPY) dyes are promising photosensitizers for cellular imaging and photodynamic therapy (PDT) owing to their excellent photophysical properties and the synthetically tunable core. Metalation provides a convenient way to overcome the drawbacks arising from their low aqueous solubility. New photo-/redox-responsive Co(III) prodrug chaperones are developed as anticancer PDT agents for efficient cellular delivery of red-light-active BODIPY dyes. The photobiological activity of heteroleptic Co(III) complexes derived from tris(2-pyridylmethyl)amine (TPA) and acetylacetone-conjugated PEGylated distyryl BODIPY (HL1) or its dibromo analogue (HL2), [CoIII(TPA)(L1/L2)](ClO4)2 (1 and 2), are investigated. The Co(III)/Co(II) redox potential is tuned using the Co(III)-TPA scaffold. Complex 1 displays the in vitro release of BODIPY on red light irradiation. Complex 2, having good singlet oxygen quantum yield (ΦΔ âˆ¼ 0.28 in DMSO), demonstrates submicromolar photocytotoxicity to HeLa cancer cells (IC50 ≈ 0.23 µM) while being less toxic to HPL1D normal cells in red light. Cellular imaging using the emissive complex 1 shows mitochondrial localization and significant penetration into the HeLa tumor spheroids. Complex 2 shows supercoiled DNA photocleavage activity and apoptotic cell death through phototriggered generation of reactive oxygen species. The Co(III)-BODIPY prodrug conjugates exemplify new type of phototherapeutic agents with better efficacy than the organic dyes alone in the phototherapeutic window.


Assuntos
Antineoplásicos , Fotoquimioterapia , Porfobilinogênio/análogos & derivados , Pró-Fármacos , Humanos , Boro/farmacologia , Luz Vermelha , Corantes , Pró-Fármacos/farmacologia , Cobalto/farmacologia , Fármacos Fotossensibilizantes/efeitos da radiação , Antineoplásicos/efeitos da radiação , Compostos de Boro/farmacologia , Compostos de Boro/efeitos da radiação , Oxigênio Singlete/metabolismo , Luz
2.
Eur J Med Chem ; 261: 115816, 2023 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-37717381

RESUMO

A combination of photodynamic therapy (PDT) and photothermal therapy (PTT) within the phototherapeutic window (600-900 nm) can lead to significantly enhanced therapeutic outcomes, surpassing the efficacy observed with PDT or PTT alone in cancer phototherapy. Herein, we report a novel small-molecule mixed-ligand Ni(II)-dithiolene complex (Ni-TDD) with a dipyridophenazine ligand, demonstrating potent red-light PDT and significant near-infrared (NIR) light mild-temperature PTT activity against cancer cells and 3D multicellular tumour spheroids (MCTSs). The four-coordinate square planar complex exhibited a moderately intense absorption band (ε âˆ¼ 3700 M-1cm-1) centered around 900 nm and demonstrated excellent dark and photostability in an aqueous phase. Ni-TDD induced a potent red-light (600-720 nm) PDT effect on HeLa cancer cells (IC50 = 1.8 µM, photo irritation factor = 44), triggering apoptotic cell death through efficient singlet oxygen generation. Ni-TDD showed a significant intercalative binding affinity towards double-helical calf thymus DNA, resulting in a binding constant (Kb) âˆ¼ 106 M-1. The complex induced mild hyperthermia and exerted a significant mild-temperature PTT effect on MDA-MB-231 cancer cells upon irradiation with 808 nm NIR light. Simultaneous irradiation of Ni-TDD-treated HeLa MCTSs with red and NIR light led to a remarkable synergistic inhibition of growth, exceeding the effects of individual irradiation, through the generation of singlet oxygen and mild hyperthermia. Ni-TDD displayed minimal toxicity towards non-cancerous HPL1D and L929 cells, even at high micromolar concentrations. This is the first report of a Ni(II) complex demonstrating red-light PDT activity and the first example of a first-row transition metal complex exhibiting combined PDT and PTT effects within the clinically relevant phototherapeutic window. Our findings pave the way for designing and developing metal-dithiolene complexes as dual-acting cancer phototherapy agents using long wavelength light for treating solid tumors.


Assuntos
Hipertermia Induzida , Neoplasias , Fotoquimioterapia , Humanos , Oxigênio Singlete , Ligantes , Hipertermia Induzida/métodos , Fotoquimioterapia/métodos , Células HeLa , Fototerapia , Fármacos Fotossensibilizantes/química , Linhagem Celular Tumoral , Neoplasias/tratamento farmacológico
3.
J Inorg Biochem ; 244: 112226, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37105008

RESUMO

To overcome the drawbacks associated with chemotherapeutic and porphyrin-based photodynamic therapy (PDT) agents, the use of BODIPY (boron-dipyrromethene) scaffold has gained prominence in designing a new generation of photosensitizers-cum-cellular imaging agents. However, their poor cell permeability and limited solubility in aqueous medium inhibits the in-vitro application of their organic form. This necessitates the development of metal-BODIPY conjugates with improved physiological stability and enhanced therapeutic efficacy. We have designed two iron(III)-BODIPY conjugates, [Fe(L1/2)(L3)Cl] derived from benzyl-dipicolylamine and its glycosylated analogue along with a BODIPY-tagged catecholate. The complexes showed intense absorption bands (ε âˆ¼ 55,000 M-1 cm-1) and demonstrated apoptotic PDT activity upon red-light irradiation (30 J/cm2, 600-720 nm). The complex with singlet oxygen quantum yield value of ∼0.34 gave sub-micromolar IC50 (half-maximal inhibitory concentration) value (∼0.08 µM) in both HeLa and H1299 cancer cells with a photocytotoxicity index value of >1200. Both the complexes were found to have significantly lower cytotoxic effects in non-cancerous HPL1D (human peripheral lung epithelial) cells. Singlet oxygen was determined to be the prime reactive oxygen species (ROS) responsible for cell damage from pUC19 DNA photo-cleavage studies, 1,3-diphenylisobenzofuran and SOSG (Singlet Oxygen Sensor Green) assays. Cellular imaging studies showed excellent fluorescence from complex 2 within 4 h, with localization in lysosomes. Significant drug accumulation into the core of 3D multicellular tumor spheroids was observed within 8 h from intense in-vitro emission. The complexes exemplify iron-based targeted PDT agents and show promising results as potential transition metal-based drugs for ROS mediated red light photocytotoxicity with low dosage requirement.


Assuntos
Antineoplásicos , Fotoquimioterapia , Humanos , Boro/farmacologia , Oxigênio Singlete , Espécies Reativas de Oxigênio , Ferro , Luz , Fármacos Fotossensibilizantes/efeitos da radiação , Compostos de Boro/farmacologia , Compostos de Boro/efeitos da radiação , Antineoplásicos/farmacologia , Lisossomos
4.
RSC Med Chem ; 13(12): 1526-1539, 2022 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-36561074

RESUMO

A cisplatin-based platinum(iv) prodrug, [Pt(NH3)2Cl2(OH)(L 1 )], having L 1 as a red-light active boron-dipyrromethene (BODIPY) pendant, was synthesized and characterized and its application as a chemo-cum-photodynamic therapy agent was studied. Me-L 1 as the ligand precursor is structurally characterized. The complex displayed an intense absorption band near 650 nm (ε ∼ 8.8 × 104 dm3 mol-1 cm-1) in 1 : 1 (v/v) DMSO/DPBS. It showed an emission band at 674 nm (λ ex = 630 nm) with a fluorescence quantum yield (Φ F) value of 0.37. In red light (600-720 nm), it generated singlet oxygen as evidenced from the 1,3-diphenylisobenzofuran (DPBF) titration experiment giving a singlet oxygen quantum yield (Φ Δ) value of 0.28 in DMSO. The mechanistic pUC19 DNA photocleavage study and singlet oxygen sensor green (SOSG) assay ascertained its ability to generate singlet oxygen in both extracellular and intracellular media by a type-II photo-process. The complex exhibited high stability in the dark, but on red-light irradiation, it displayed rapid activation in the presence of a reducing environment. It displayed remarkable apoptotic photocytotoxicity with half-maximal inhibitory concentration (IC50) ranging from 0.58 to 0.76 µM in human cervical cancer (HeLa) and breast cancer (MCF-7) cells with a respective photo-cytotoxicity index value of >172 and >131. The photodynamic activity was significantly less in non-cancerous human peripheral lung epithelial (HPL1D) cells. The emissive complex showed localization in the mitochondria and endoplasmic reticulum (ER) with a similar Pearson's correlation coefficient value, making it a dual organelle-targeted therapeutic agent. JC-1, fluo-4-AM and annexin V-FITC/propidium iodide assays in HeLa cells showed cellular apoptosis by arresting cells in the sub-G1 phase via mitochondrial dysfunction and ER stress.

5.
Dalton Trans ; 51(27): 10392-10405, 2022 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-35758169

RESUMO

Two multichromophoric homoleptic ruthenium(II) complexes [Ru(tpy-BODIPY)2]Cl2 (complexes 1 and 2, tpy = 4-phenyl-2,2:6,2-terpyridine, BODIPY = boron-dipyrromethene) were prepared, characterized and their phototherapeutic activity and bioimaging properties were studied. The complexes having structural similarity differ only by a phenylethynyl linker, and its overall influence on their physicochemical and photobiological behavior was evaluated. The terpyridine-BODIPY ligand L1 was structurally characterized by X-ray crystallography. The complexes showed intense absorption near 500 nm (ε: ∼1.5 × 105 M-1 cm-1 in DMSO), have a high singlet oxygen quantum yield (ΦΔ: ∼0.6 in DMSO), and displayed low photobleaching thus making them suitable for PDT applications. The complexes showed high DNA binding affinity and induced DNA damage on light activation via multiple types of ROS production. Confocal laser scanning microscopy experiments revealed their incorporation in the cancer cells and complex 1 predominantly accumulated in lysosomes. The complexes displayed a significant PDT effect in cancerous cells with visible light activation with a high photocytotoxicity index (PI) value in HeLa cells. Both type-I and type-II photosensitization processes were involved in the PDT effect. The photodynamic action of complex 2 initiated cellular apoptosis. Finally, their diagnostic potential was evaluated against clinically relevant 3D multicellular tumor spheroids (MCTs).


Assuntos
Complexos de Coordenação , Fotoquimioterapia , Rutênio , Compostos de Boro , Complexos de Coordenação/química , Complexos de Coordenação/farmacologia , Dimetil Sulfóxido , Células HeLa , Humanos , Luz , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Rutênio/química , Rutênio/farmacologia
6.
Inorg Chem ; 60(9): 6649-6662, 2021 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-33855849

RESUMO

The bioessential nature of cobalt and the rich photochemistry of its coordination complexes can be exploited to develop potential next-generation photochemotherapeutics. A series of six novel mixed-ligand cobalt(III) complexes of the formulation [Co(B)2(L)]ClO4 (1-6), where B is an N,N-donor phenanthroline base, namely, 1,10-phenanthroline (phen in 1 and 4), dipyrido[3,2-d:2',3'-f]quinoxaline (dpq in 2 and 5), and dipyrido[3,2-a:2',3'-c]phenazine (dppz in 3 and 6), and L is an O,O-donor dianionic ligand derived from catechol (1,2-dihydroxybenzene, cat2-, in 1-3) or esculetin (6,7-dihydoxycoumarin, esc2-, in 4-6), have been prepared and characterized, and their light-triggered cytotoxicity has been studied in cancer cells. The single-crystal X-ray diffraction structures of complexes 1 (as PF6- salt, 1a) and 2 show distorted octahedral geometries around the cobalt(III) center formed by the set of N4O2 donor atoms. The low-spin and 1:1 electrolytic complexes 1-6 display a d-d transition around 700 nm. Complexes 4-6 with a coordinated esc2- ligand additionally display a π → π* intraligand transition centered at 403 nm. Complexes 4-6 possessing a naturally occurring and photoactive esc2- ligand show high visible-light-triggered cytotoxicity against HeLa and MCF-7 cancer cells, yielding remarkably low micromolar IC50 values while being much less toxic under dark conditions. Control complexes 1-3 possessing the photoinactive cat2- ligand show significantly less cytotoxicity either in the presence of light or in the dark. The complex-induced cell death is apoptotic in nature caused by the formation of reactive oxygen species via a type 1 photoredox pathway. Fluorescence microscopy of HeLa cells treated with complex 6 reveals mitochondrial localization of the complex. A significant decrease in the dark toxicity of free esculetin and dppz base is observed upon coordination to cobalt(III). Complexes bind to calf-thymus DNA with significant affinity, but 6 binds with the greatest affinity. Complex 6 efficiently photocleaves supercoiled DNA to its nicked circular form when irradiated with visible light via a photoredox type 1 pathway involving hydroxyl radicals (HO•). Thus, complex 6 showing remarkable visible-light-triggered cytotoxicity but negligible toxicity in the dark is a good candidate for cancer photochemotherapy applications.


Assuntos
Antineoplásicos/farmacologia , Complexos de Coordenação/farmacologia , Mitocôndrias/efeitos dos fármacos , Fotoquímica , Antineoplásicos/síntese química , Antineoplásicos/química , Proliferação de Células/efeitos dos fármacos , Cobalto/química , Cobalto/farmacologia , Complexos de Coordenação/síntese química , Complexos de Coordenação/química , Cumarínicos/química , Cumarínicos/farmacologia , Cristalografia por Raios X , Teoria da Densidade Funcional , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Ligantes , Mitocôndrias/metabolismo , Modelos Moleculares , Estrutura Molecular , Fenantrolinas/química , Fenantrolinas/farmacologia , Células Tumorais Cultivadas
7.
ACS Omega ; 5(8): 4282-4292, 2020 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-32149258

RESUMO

Mixed-ligand oxidovanadium(IV) ß-diketonates having NNN-donor dipicolylamine-conjugated to boron-dipyrromethene (BODIPY in L1) and diiodo-BODIPY (in L2) moieties, namely, [VO(L1)(acac)]Cl (1), [VO(L2)(acac)]Cl (2), and [VO(L1)(dbm)]Cl (3), where acac and dbm are monoanionic O,O-donor acetylacetone and 1,3-diphenyl-1,3-propanedione, were prepared, characterized, and tested for their photoinduced anticancer activity in visible light. Complexes 1 and 2 were structurally characterized as their PF6 - salts (1a and 2a) by X-ray crystallography. They showed VIVN3O3 six-coordinate geometry with dipicolylamine base as the facial ligand. The non-iodinated BODIPY complexes displayed absorption maxima at ∼501 nm, while it is ∼535 nm for the di-iodinated 2 in 10% DMSO-PBS buffer medium (pH = 7.2). Complexes 1 and 3 being green emissive (λem, ∼512 nm; λex, 470 nm; ΦF, ∼0.10) in 10% aqueous DMSO were used for cellular imaging studies. Complex 3 localized primarily in the mitochondria of the cervical HeLa cells with a co-localization coefficient value of 0.7. The non-emissive diiodo-BODIPY complex 2 showed generation of singlet oxygen (ΦΔ ≈ 0.47) on light activation. Annexin-V assay showed singlet oxygen-mediated cellular apoptosis, making this complex a targeted PDT agent.

8.
J Inorg Biochem ; 202: 110817, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31706182

RESUMO

Cis-dichloro-oxovanadium(IV) complexes [VO(L1/L2)Cl2], where L1 is N-(4-(5,5-difluoro-1,3,7,9-tetramethyl-5H-4ʎ4,5ʎ4-dipyrrolo[1,2-c:2',1'-f][1,3,2]diazaborinin-10-yl)benzyl)-1-(pyridin-2-yl)-N-(pyridin-2-ylmethyl)methanamine in 1 and L2 is N-(4-(5,5-difluoro-2,8-diiodo-1,3,7,9-tetramethyl-5H-4ʎ4,5ʎ4-dipyrrolo[1,2-c:2',1'-f][1,3,2]diazaborinin-10-yl)benzyl)-1-(pyridin-2-yl)-N-(pyridin-2-ylmethyl)methanamine in 2) having 4,4-difluoro-4-bora-3a,4a-diaza-s-indacene as boron-dipyrromethene (BODIPY) appended dipicolylamine bases were prepared, characterized and their photocytotoxicity studied. X-ray crystal structure of 1 showed distorted octahedral geometry with a VIVON3Cl2 core having Cl-V-Cl angle of 91.93(4)°. The complexes showed variable solution conductivity properties. They were non-electrolytes in dry DMF at 25 °C but showed 1:1 electrolytic behavior in an aqueous medium due to dissociation of one chloride ligand as evidenced from the mass spectral study. Complexes 1 and 2 showed absorption bands at 500 and 535 nm, respectively. The calf thymus DNA melting study revealed their interaction through DNA crosslinking on exposure to light which was further confirmed from the alkaline agarose gel electrophoresis using plasmid supercoiled pUC19 DNA. Complex 2 showed disruption of the mitochondrial membrane potential in the JC-1 (1,1',3,3'-tetraethyl-5,5',6,6'-tetrachloroimidacarbocyanine iodide) assay. The complexes were photocytotoxic in visible light (400-700 nm, power: 10 J cm-2) in cervical cancer HeLa and breast cancer MCF-7 cells. Complex 2 having a photoactive diiodo­boron-dipyrromethene moiety gave a singlet oxygen quantum yield (ΦΔ) value of ~0.6. It showed singlet oxygen mediated apoptotic photodynamic therapy activity with remarkably low IC50 (half maximal inhibitory concentration) value of ~0.15 µM. The cis-disposition of chlorides gave a cis-divacant 4-coordinate intermediate structure from the density functional theory (DFT) study thus mimicking the DNA crosslinking property of cisplatin.


Assuntos
Compostos de Boro , Citotoxinas , DNA , Fármacos Fotossensibilizantes , Porfobilinogênio/análogos & derivados , Vanadatos , Boro/química , Boro/farmacologia , Compostos de Boro/síntese química , Compostos de Boro/química , Compostos de Boro/farmacologia , Cristalografia por Raios X , Citotoxinas/síntese química , Citotoxinas/química , Citotoxinas/farmacologia , DNA/química , DNA/metabolismo , Células HeLa , Humanos , Células MCF-7 , Fármacos Fotossensibilizantes/síntese química , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Porfobilinogênio/síntese química , Porfobilinogênio/química , Porfobilinogênio/farmacologia , Vanadatos/química , Vanadatos/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA