Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Am Chem Soc ; 143(48): 20224-20240, 2021 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-34808054

RESUMEN

The Pt(IV) prodrug trans, trans, trans-[Pt(pyridine)2(N3)2(OH)2] (Pt1) and its coumarin derivative trans, trans, trans-[Pt(pyridine)2(N3)2(OH)(coumarin-3-carboxylate)] (Pt2) are promising agents for photoactivated chemotherapy. These complexes are inert in the dark but release Pt(II) species and radicals upon visible light irradiation, resulting in photocytotoxicity toward cancer cells. Here, we have used synchrotron techniques to investigate the in-cell behavior of these prodrugs and visualize, for the first time, changes in cellular morphology and Pt localization upon treatment with and without light irradiation. We show that photoactivation of Pt2 induces remarkable cellular damage with extreme alterations to multiple cellular components, including formation of vacuoles, while also significantly increasing the cellular accumulation of Pt species compared to dark conditions. X-ray absorption near-edge structure (XANES) measurements in cells treated with Pt2 indicate only partial reduction of the prodrug upon irradiation, highlighting that phototoxicity in cancer cells may involve not only Pt(II) photoproducts but also photoexcited Pt(IV) species.


Asunto(s)
Antineoplásicos/farmacología , Complejos de Coordinación/farmacología , Profármacos/farmacología , Antineoplásicos/química , Antineoplásicos/efectos de la radiación , Proliferación Celular/efectos de los fármacos , Complejos de Coordinación/química , Complejos de Coordinación/efectos de la radiación , Humanos , Luz , Células PC-3 , Platino (Metal)/química , Platino (Metal)/efectos de la radiación , Profármacos/química , Profármacos/efectos de la radiación , Análisis de la Célula Individual
2.
Nat Chem ; 13(12): 1248-1256, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34697400

RESUMEN

Companion diagnostics (CDx) are powerful tests that can provide physicians with crucial biomarker information that can improve treatment outcomes by matching therapies to patients. Here, we report a photoacoustic imaging-based CDx (PACDx) for the selective detection of elevated glutathione (GSH) in a lung cancer model. GSH is abundant in most cells, so we adopted a physical organic chemistry approach to precisely tune the reactivity to distinguish between normal and pathological states. To evaluate the efficacy of PACDx in vivo, we designed a blind study where photoacoustic imaging was used to identify mice bearing lung xenografts. We also employed PACDx in orthotopic lung cancer and liver metastasis models to image GSH. In addition, we designed a matching prodrug, PARx, that uses the same SNAr chemistry to release a chemotherapeutic with an integrated PA readout. Studies demonstrate that PARx can inhibit tumour growth without off-target toxicity in a lung cancer xenograft model.


Asunto(s)
Arilsulfonatos/química , Biomarcadores de Tumor/metabolismo , Colorantes/química , Glutatión/metabolismo , Indoles/química , Neoplasias Pulmonares/tratamiento farmacológico , Animales , Arilsulfonatos/síntesis química , Arilsulfonatos/efectos de la radiación , Línea Celular Tumoral , Colorantes/síntesis química , Colorantes/efectos de la radiación , Desoxicitidina/análogos & derivados , Desoxicitidina/síntesis química , Desoxicitidina/efectos de la radiación , Desoxicitidina/uso terapéutico , Diseño de Fármacos , Femenino , Células HEK293 , Humanos , Indoles/síntesis química , Indoles/efectos de la radiación , Luz , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Técnicas Fotoacústicas/métodos , Profármacos/síntesis química , Profármacos/efectos de la radiación , Profármacos/uso terapéutico , Método Simple Ciego , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
3.
J Inorg Biochem ; 223: 111526, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34246120

RESUMEN

Oxoplatin-B, a platinum(IV) complex [Pt(NH3)2Cl2(L1)(OH)] (1) of 4-methylbenzoic acid (HL1) functionalized with 4,4-difluoro-4-bora-3a,4a-diaza-s-indacene (BODIPY) was prepared, characterized and its antitumor activity studied. [Pt(NH3)2Cl2(L2)(OH)] (2) of 4-methylbenzoic acid (HL2) was studied as a control. Complex 1 showed an absorption band at 500 nm (ɛ = 4.34 × 104 M-1 cm-1) and an emission band at 515 nm (λex = 488 nm, ΦF = 0.64) in 1% dimethyl sulfoxide/Dulbecco's Modified Eagle's Medium (pH = 7.2). Visible light-induced (400-700 nm) generation of singlet oxygen was evidenced from 1,3-diphenylisobenzofuran titration study. Complex 1 showed photo-induced cytotoxicity in visible light (400-700 nm, 10 J cm-2) against human breast cancer (MCF-7), cervical cancer (HeLa) and lung cancer (A549) cells (IC50: 1.1-3.8 µM) while being less toxic in normal cells. Confocal imaging showed mitochondrial localization with additional evidence from platinum content from isolated mitochondria and 5,5,6,6'-tetrachloro-1,1',3,3' tetraethylbenzimi-dazoylcarbocyanine iodide (JC-1) assay. Cellular apoptosis was observed from Annexin-V-FITC (fluorescein isothiocyanate)/propidium iodide assay.


Asunto(s)
Antineoplásicos/farmacología , Complejos de Coordinación/farmacología , Mitocondrias/efectos de los fármacos , Fármacos Fotosensibilizantes/farmacología , Profármacos/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/efectos de la radiación , Apoptosis/efectos de los fármacos , Benzoatos/síntesis química , Benzoatos/farmacología , Benzoatos/efectos de la radiación , Compuestos de Boro/síntesis química , Compuestos de Boro/farmacología , Compuestos de Boro/efectos de la radiación , Bovinos , Línea Celular Tumoral , Complejos de Coordinación/síntesis química , Complejos de Coordinación/efectos de la radiación , ADN/efectos de los fármacos , División del ADN/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Luz , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Fotoquimioterapia , Fármacos Fotosensibilizantes/síntesis química , Fármacos Fotosensibilizantes/efectos de la radiación , Platino (Metal)/química , Platino (Metal)/efectos de la radiación , Profármacos/síntesis química , Profármacos/efectos de la radiación , Oxígeno Singlete/metabolismo
4.
Nat Chem ; 13(8): 805-810, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34112990

RESUMEN

Chemotherapy is a powerful tool in the armoury against cancer, but it is fraught with problems due to its global systemic toxicity. Here we report the proof of concept of a chemistry-based strategy, whereby gamma/X-ray irradiation mediates the activation of a cancer prodrug, thereby enabling simultaneous chemo-radiotherapy with radiotherapy locally activating a prodrug. In an initial demonstration, we show the activation of a fluorescent probe using this approach. Expanding on this, we show how sulfonyl azide- and phenyl azide-caged prodrugs of pazopanib and doxorubicin can be liberated using clinically relevant doses of ionizing radiation. This strategy is different to conventional chemo-radiotherapy radiation, where chemo-sensitization of the cancer takes place so that subsequent radiotherapy is more effective. This approach could enable site-directed chemotherapy, rather than systemic chemotherapy, with 'real time' drug decaging at the tumour site. As such, it opens up a new era in targeted and directed chemotherapy.


Asunto(s)
Azidas/uso terapéutico , Neoplasias/tratamiento farmacológico , Profármacos/uso terapéutico , Animales , Antineoplásicos/química , Antineoplásicos/efectos de la radiación , Antineoplásicos/uso terapéutico , Azidas/química , Azidas/efectos de la radiación , Doxorrubicina/análogos & derivados , Doxorrubicina/efectos de la radiación , Doxorrubicina/uso terapéutico , Femenino , Colorantes Fluorescentes/química , Colorantes Fluorescentes/efectos de la radiación , Rayos gamma , Células HeLa , Células Endoteliales de la Vena Umbilical Humana , Humanos , Indazoles/química , Indazoles/efectos de la radiación , Indazoles/uso terapéutico , Ratones Endogámicos BALB C , Ratones Desnudos , Oxidación-Reducción , Profármacos/química , Profármacos/efectos de la radiación , Prueba de Estudio Conceptual , Pirimidinas/química , Pirimidinas/efectos de la radiación , Pirimidinas/uso terapéutico , Sulfonamidas/química , Sulfonamidas/efectos de la radiación , Sulfonamidas/uso terapéutico , Rayos X , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Inorg Biochem ; 222: 111508, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34116426

RESUMEN

Histone deacetylase inhibitors have often been used in combination treatment of various types of cancers due to their non-genotoxic epigenetic potential. Valproic acid (VPA) is a well-known histone deacetylase inhibitor. Conjugate of VPA with a phtoactive platinum diimine complex through an ester bond has been fabricated to potentiate the photocytotoxicity of the photosensitizer. Its capability to generate singlet oxygen, behavior in the presence of esterase, and photocytotoxicity in tumor cells have also been studied. The results revealed that the novel VPA-modified platinum diimine complex could produce singlet oxygen efficiently and release VPA in the presence of porcine liver esterase. The results also suggested that incorporation of VPA moiety into the platinum diimine complex might significantly enhance the cytotoxicity of the complex.


Asunto(s)
Complejos de Coordinación/farmacología , Fármacos Fotosensibilizantes/farmacología , Ácido Valproico/análogos & derivados , Ácido Valproico/farmacología , Animales , Hidrolasas de Éster Carboxílico/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Complejos de Coordinación/síntesis química , Complejos de Coordinación/metabolismo , Complejos de Coordinación/efectos de la radiación , Inhibidores de Histona Desacetilasas/síntesis química , Inhibidores de Histona Desacetilasas/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/efectos de la radiación , Humanos , Luz , Fotoquimioterapia , Fármacos Fotosensibilizantes/síntesis química , Fármacos Fotosensibilizantes/metabolismo , Fármacos Fotosensibilizantes/efectos de la radiación , Platino (Metal)/química , Profármacos/síntesis química , Profármacos/metabolismo , Profármacos/farmacología , Profármacos/efectos de la radiación , Oxígeno Singlete/metabolismo , Porcinos , Ácido Valproico/metabolismo , Ácido Valproico/efectos de la radiación
6.
ACS Chem Biol ; 16(7): 1128-1141, 2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-34114796

RESUMEN

Hydrogen sulfide (H2S) has gained significant attention as a potent bioregulator in the redox metabolome, but it is just one of many reactive sulfur species (RSS). Recently, small molecule persulfides (structure RSSH) have emerged as RSS of particular interest due to their enhanced antioxidant abilities compared to H2S and their ability to directly convert protein thiols into protein persulfides, suggesting that persulfides may have distinct physiological functions from H2S. However, persulfides exhibit instability and cross-reactivity that hampers the elucidation of their precise biological roles. As such, chemists have designed chemical tools and techniques to facilitate the study of persulfides under various conditions. These molecules and methods include persulfide trapping reagents and sensors, as well as compounds that degrade in response to various triggers to release persulfides, termed persulfide donors. There now exist a variety of persulfide donor classes, some of which possess tissue-targeting capabilities designed to mimic localized endogenous production of RSS. This Review briefly covers the physicochemical properties of persulfides, the endogenous production of small molecule persulfides, and their reactions with protein thiols and other reactive species. These introductory sections are followed by a discussion of chemical tools used in persulfide chemical biology, with critical analysis of recent advancements in the field and commentary on potential directions for future research.


Asunto(s)
Sulfuros/análisis , Sulfuros/farmacología , Animales , Línea Celular Tumoral , Técnicas de Química Analítica , Disulfuros/química , Disulfuros/metabolismo , Disulfuros/efectos de la radiación , Humanos , Indicadores y Reactivos/química , Luz , Oxidación-Reducción , Profármacos/química , Profármacos/metabolismo , Profármacos/farmacología , Profármacos/efectos de la radiación , Sulfuros/química , Sulfuros/metabolismo
7.
Theranostics ; 11(7): 3502-3511, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33537100

RESUMEN

Photodynamic therapy (PDT) has emerged as one of the most up-and-coming non-invasive therapeutic modalities for cancer therapy in rencent years. However, its therapeutic effect was still hampered by the short life span, limited diffusion distance and ineluctable depletion of singlet oxygen (1O2), as well as the hypoxic microenvironment in the tumor tissue. Such problems have limited the application of PDT and appropriate solutions are highly demand. Methods: Herein, a programmatic treatment strategy is proposed for the development of a smart molecular prodrug (D-bpy), which comprise a two-photon photosensitizer and a hypoxia-activated chemotherapeutic prodrug. A rhodamine dye was designed to connect them and track the drug release by the fluorescent signal generated through azo bond cleavage. Results: The prodrug (D-bpy) can stay on the cell membrane and enrich at the tumor site. Upon light irradiation, the therapeutic effect was enhanced by a stepwise treatment: (i) direct generation of 1O2 on the cell membrane induced membrane destruction and promoted the D-bpy uptake; (ii) deep tumor hypoxia caused by two-photon PDT process further triggered the activation of the chemotherapy prodrug. Both in vitro and in vivo experiments, D-bpy have exhabited excellent tumor treatment effect. Conclusion: The innovative programmatic treatment strategy provides new strategy for the design of follow-up anticancer drugs.


Asunto(s)
Hipoxia/tratamiento farmacológico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Fotoquimioterapia/métodos , Fotones , Fármacos Fotosensibilizantes/farmacología , Profármacos/farmacología , Animales , Compuestos Azo/química , Membrana Celular/patología , Membrana Celular/efectos de la radiación , Femenino , Colorantes Fluorescentes/química , Células HeLa , Humanos , Hipoxia/metabolismo , Hipoxia/patología , Neoplasias Mamarias Experimentales/química , Neoplasias Mamarias Experimentales/patología , Fármacos Fotosensibilizantes/síntesis química , Fármacos Fotosensibilizantes/efectos de la radiación , Profármacos/síntesis química , Profármacos/efectos de la radiación , Rodaminas/química , Oxígeno Singlete/química , Coloración y Etiquetado/métodos
8.
Eur J Med Chem ; 215: 113251, 2021 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-33611187

RESUMEN

Development of the drug with high therapeutic efficacy and low toxicity is crucial to cancer ablation. In this study, we have demonstrated a red light-responsive prodrug BDP-TK-CPT by connecting the chemotherapeutic agent camptothecin with a boron dipyrromethene (BDP)-based photosensitizer via a reactive oxygen species (ROS)-labile thioketal chain. Since camptothecin is modified by a BDP-based macrocycle at the active site, the formed prodrug displays an extremely low toxicity in dark. However, upon illumination by red light, it can efficiently generate ROS leading to cell death by photodynamic therapy. Meanwhile, the ROS generated can destroy thioketal group to release free camptothecin which further results in local cell death by chemotherapy. The combined antitumor effects of the prodrug have been verified in HepG2, EC109, and HeLa cancer cells and mice bearing H22 tumors. This study may provide an alternative strategy for stimuli-responsive combination treatment of tumors by conjugation of ROS-activatable prodrugs with photosensitizing agents.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Fármacos Fotosensibilizantes/uso terapéutico , Profármacos/uso terapéutico , Animales , Antineoplásicos/síntesis química , Antineoplásicos/efectos de la radiación , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Terapia Combinada , Quimioterapia , Femenino , Humanos , Luz , Ratones , Necrosis/inducido químicamente , Fotoquimioterapia , Fármacos Fotosensibilizantes/síntesis química , Fármacos Fotosensibilizantes/efectos de la radiación , Profármacos/síntesis química , Profármacos/efectos de la radiación , Especies Reactivas de Oxígeno/metabolismo
9.
Adv Drug Deliv Rev ; 171: 94-107, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33486009

RESUMEN

Light is a uniquely powerful tool for controlling molecular events in biology. No other external input (e.g., heat, ultrasound, magnetic field) can be so tightly focused or so highly regulated as a clinical laser. Drug delivery vehicles that can be photonically activated have been developed across many platforms, from the simplest "caging" of therapeutics in a prodrug form, to more complex micelles and circulating liposomes that improve drug uptake and efficacy, to large-scale hydrogel platforms that can be used to protect and deliver macromolecular agents including full-length proteins. In this Review, we discuss recent innovations in photosensitive drug delivery and highlight future opportunities to engineer and exploit such light-responsive technologies in the clinical setting.


Asunto(s)
Sistemas de Liberación de Medicamentos , Luz , Animales , Materiales Biocompatibles/administración & dosificación , Materiales Biocompatibles/efectos de la radiación , Preparaciones de Acción Retardada/administración & dosificación , Preparaciones de Acción Retardada/efectos de la radiación , Humanos , Liposomas/administración & dosificación , Liposomas/efectos de la radiación , Micelas , Profármacos/administración & dosificación , Profármacos/efectos de la radiación
10.
Org Biomol Chem ; 19(8): 1752-1759, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33355577

RESUMEN

Recently, sunscreen-based drug photocages have been introduced to provide UV protection to photoactive drugs, thus increasing their photosafety. Here, combined experimental and theoretical studies performed on a photocage based on the commercial UVA filter avobenzone (AB) and on the photosensitizing non-steroidal anti-inflammatory drug ketoprofen (KP) are presented unveiling the photophysical processes responsible for the light-triggered release. Particular attention is paid to solvent stabilization of the drug and UV filter excited states, respectively, which leads to a switching between the triplet excited state energies of the AB and KP units. Most notably, we show that the stabilization of the AB triplet excited state in ethanol solution is the key requirement for an efficient photouncaging. By contrast, in apolar solvents, in particular hexane, KP has the lowest triplet excited state, hence acting as an energy acceptor quenching the AB triplet manifold, thus inhibiting the desired photoreaction.


Asunto(s)
Antiinflamatorios no Esteroideos/química , Cetoprofeno/química , Fármacos Fotosensibilizantes/química , Profármacos/química , Propiofenonas/química , Protectores Solares/química , Antiinflamatorios no Esteroideos/efectos de la radiación , Etanol/química , Hexanos/química , Cetoprofeno/efectos de la radiación , Modelos Químicos , Fotólisis/efectos de la radiación , Fármacos Fotosensibilizantes/efectos de la radiación , Profármacos/efectos de la radiación , Propiofenonas/efectos de la radiación , Solventes/química , Protectores Solares/efectos de la radiación , Rayos Ultravioleta
11.
Chemistry ; 26(48): 10992-11006, 2020 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-32700815

RESUMEN

Two photoactivatable dicarbonyl ruthenium(II) complexes based on an amide-functionalised bipyridine scaffold (4-position) equipped with an alkyne functionality or a green-fluorescent BODIPY (boron-dipyrromethene) dye have been prepared and used to investigate their light-induced decarbonylation. UV/Vis, FTIR and 13 C NMR spectroscopies as well as gas chromatography and multivariate curve resolution alternating least-squares analysis (MCR-ALS) were used to elucidate the mechanism of the decarbonylation process. Release of the first CO molecule occurs very quickly, while release of the second CO molecule proceeds more slowly. In vitro studies using two cell lines A431 (human squamous carcinoma) and HEK293 (human embryonic kidney cells) have been carried out in order to characterise the anti-proliferative and anti-apoptotic activities. The BODIPY-labelled compound allows for monitoring the cellular uptake, showing fast internalisation kinetics and accumulation at the endoplasmic reticulum and mitochondria.


Asunto(s)
2,2'-Dipiridil/química , Monóxido de Carbono/química , Profármacos/química , Profármacos/efectos de la radiación , Rutenio/química , Línea Celular Tumoral , Células HEK293 , Humanos
12.
Biomater Sci ; 8(11): 3116-3129, 2020 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-32352102

RESUMEN

To enhance the specificity and efficiency of anti-tumor therapies, we have designed a multifunctional nanoparticle platform for photochemotherapy using fluorescence (FL) and photoacoustic (PA) imaging guidance. Nanoparticles (NPs) composed of a eutectic mixture of natural fatty acids that undergo a solid-liquid phase transition at 39 °C were used to encapsulate materials for the rapid and uniform release of the hypoxia-activated prodrug tirapazamine (TPZ) and the photosensitizer IR780, which targets the mitochondria of tumor cells and can be used to induce hypoxic cell death via photodynamic therapy and photothermal therapy. In vitro, the NPs containing TPZ and IR7890 exhibited appreciable cell uptake and triggered drug release when irradiated with a NIR laser. In vivo, photochemotherapy of the NPs achieved the best anti-tumor efficacy under PA and FL imaging guidance and monitoring. By combining IR780 mitochondria-targeting phototherapy with TPZ, we observed improved anti-tumor effectiveness and this has the potential to reduce the side effects of traditional chemotherapy. Herein, we demonstrate a new intracellular photochemotherapy nanosystem that co-encapsulates photosensitizers and hypoxia-activated drugs to enhance the overall anti-tumor effect precisely and efficiently.


Asunto(s)
Antineoplásicos/administración & dosificación , Indoles/administración & dosificación , Nanopartículas/administración & dosificación , Neoplasias/tratamiento farmacológico , Fotoquimioterapia , Fármacos Fotosensibilizantes/administración & dosificación , Profármacos/administración & dosificación , Tirapazamina/administración & dosificación , Animales , Antineoplásicos/química , Antineoplásicos/efectos de la radiación , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Liberación de Fármacos , Femenino , Indoles/química , Indoles/efectos de la radiación , Rayos Láser , Ratones Endogámicos BALB C , Nanopartículas/química , Nanopartículas/efectos de la radiación , Neoplasias/metabolismo , Neoplasias/patología , Imagen Óptica , Técnicas Fotoacústicas , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/efectos de la radiación , Profármacos/química , Profármacos/efectos de la radiación , Especies Reactivas de Oxígeno/metabolismo , Tirapazamina/química , Tirapazamina/efectos de la radiación
13.
Chembiochem ; 20(22): 2876-2882, 2019 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-31102568

RESUMEN

Cancer is currently one of the deadliest diseases worldwide. Based on the high incidence of this disease, the side effects associated with current chemotherapies and the appearance of drug resistance, considerable efforts have been directed towards the development of new anticancer drugs with new modes of action. Metal-based compounds are particularly attractive candidates due to their metabolic mechanisms, which differ substantially from those of organic drugs. Of special interest in this context are organometallic ruthenium(II) complexes of the type [Ru(η6 -arene)(pta)Cl2 ] (arene: p-cymene, toluene, benzene, etc.; pta: 1,3,5-triaza-7-phosphaadamantane), which are abbreviated to RAPTA. Complementary to chemotherapy, photoactivated chemotherapy is a technique that has received increasing attention towards the development of treatment for numerous kinds of cancer. With this in mind, a photoactive RAPTA-type complex bearing azide ligands has been designed. The diazide complex, [Ru(η6 -p-cymene)pta-(N3 )2 ], is inert in water, but slowly releases the azide ligand upon exposure to light. Consequently, the in vitro cytotoxicity of the complex in the dark and upon light exposure at λ=450 nm in human cervical carcinoma (HeLa) and noncancerous retinal pigment epithelium (RPE-1) cells was investigated. Although the cytotoxicity of the complex was found to be modest in the dark, an increase in toxicity upon light exposure was observed.


Asunto(s)
Antineoplásicos/farmacología , Complejos de Coordinación/farmacología , Profármacos/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/efectos de la radiación , Antineoplásicos/toxicidad , Complejos de Coordinación/síntesis química , Complejos de Coordinación/efectos de la radiación , Complejos de Coordinación/toxicidad , Ensayos de Selección de Medicamentos Antitumorales , Células Epiteliales/efectos de los fármacos , Células HeLa , Humanos , Profármacos/síntesis química , Profármacos/efectos de la radiación , Profármacos/toxicidad , Epitelio Pigmentado de la Retina/citología , Rutenio/química , Rayos Ultravioleta
14.
Clin Oncol (R Coll Radiol) ; 31(5): 290-302, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30853148

RESUMEN

The role of hypoxia in radiation resistance is well established and many approaches to overcome hypoxia in tumours have been explored, with variable success. Two small molecule strategies for targeting hypoxia have dominated preclinical and clinical efforts. One approach has been the use of electron-affinic nitroheterocycles as oxygen-mimetic sensitisers. These agents are best exemplified by the 5-nitroimidazole nimorazole, which has limited use in conjunction with radiotherapy in head and neck squamous cell carcinoma. The second approach seeks to leverage tumour hypoxia as a tumour-specific address for hypoxia-activated prodrugs. These prodrugs are selectively activated by reductases under hypoxia to release cytotoxins, which in some instances may diffuse to kill surrounding oxic tumour tissue. A number of these hypoxia-activated prodrugs have been examined in clinical trial and the merits and shortcomings of recent examples are discussed. There has been an evolution from delivering DNA-interactive cytotoxins to molecularly targeted agents. Efforts to implement these strategies clinically continue today, but success has been elusive. Several issues have been identified that compromised these clinical campaigns. A failure to consider the extravascular transport and the micropharmacokinetic properties of the prodrugs has reduced efficacy. One key element for these 'targeted' approaches is the need to co-develop biomarkers to identify appropriate patients. Hypoxia-activated prodrugs require biomarkers for hypoxia, but also for appropriate activating reductases in tumours, as well as markers of intrinsic sensitivity to the released drug. The field is still evolving and changes in radiation delivery and the impact of immune-oncology will provide fertile ground for future innovation.


Asunto(s)
Hipoxia de la Célula/efectos de la radiación , Neoplasias/radioterapia , Profármacos/efectos de la radiación , Humanos
15.
J Med Chem ; 62(4): 1959-1970, 2019 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-30703330

RESUMEN

Natural lipid nanocarriers, exosomes, carry cell-signaling materials such as DNA and RNA for intercellular communications. Exosomes derived from cancer cells contribute to the progression and metastasis of cancer cells by transferring oncogenic signaling molecules to neighboring and remote premetastatic sites. Therefore, applying the unique properties of exosomes for cancer therapy has been expected in science, medicine, and drug discovery fields. Herein, we report that an exosome-targeting prodrug system, designated MARCKS-ED-photodoxaz, could spatiotemporally control the activation of an exquisitely cytotoxic agent, doxazolidine (doxaz), with UV light. The MARCKS-ED peptide enters a cell by forming a complex with the exosomes in situ at its plasma membrane and in the media. MARCKS-ED-photodoxaz releases doxaz under near-UV irradiation to inhibit cell growth with low nanomolar IC50 values. The MARCKS-ED-photodoxaz system targeting exosomes and utilizing photochemistry will potentially provide a new approach for the treatment of cancer, especially for highly progressive and invasive metastatic cancers.


Asunto(s)
Antineoplásicos/farmacología , Doxorrubicina/análogos & derivados , Exosomas/efectos de los fármacos , Nitrobencenos/farmacología , Oxazoles/farmacología , Profármacos/farmacología , Secuencia de Aminoácidos , Antineoplásicos/síntesis química , Antineoplásicos/efectos de la radiación , Línea Celular Tumoral , Membrana Celular/metabolismo , Péptidos de Penetración Celular/síntesis química , Péptidos de Penetración Celular/farmacología , Reactivos de Enlaces Cruzados/síntesis química , Reactivos de Enlaces Cruzados/farmacología , Reactivos de Enlaces Cruzados/efectos de la radiación , Doxorrubicina/síntesis química , Doxorrubicina/farmacología , Doxorrubicina/efectos de la radiación , Humanos , Nitrobencenos/síntesis química , Nitrobencenos/efectos de la radiación , Oxazoles/síntesis química , Oxazoles/efectos de la radiación , Fotólisis , Profármacos/síntesis química , Profármacos/efectos de la radiación , Rayos Ultravioleta
16.
Langmuir ; 35(5): 1919-1926, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30204452

RESUMEN

An adamantane-containing zwitterionic copolymer poly(2-(methacryloyloxy)ethyl phosphorylcholine)- co-poly(2-(methacryloyloxy)ethyl adamantane-1-carboxylate) (poly(MPC- co-MAda)) was prepared by reversible addition-fragmentation chain transfer (RAFT) polymerization. The hydrophobic photosensitizer chlorin e6 (Ce6) was conjugated to ß-cyclodextrin (ß-CD) by glutathione (GSH)-sensitive disulfide bonds. The Ce6 conjugated supramolecular prodrug nanocarriers were fabricated due to the host-guest interaction between adamantane and ß-CD, which was confirmed by dynamic light scattering (DLS) and transmission electron microscopy (TEM). The Ce6 conjugated prodrug nanocarriers showed reduction-responsive release of Ce6, which could result in the activation of Ce6. The generation of cytotoxic reactive oxygen species (ROS) was significantly enhanced due to the activation of Ce6. In additiona, the Ce6 conjugated prodrug nanocarriers could effectively inhibit the proliferation of cancer cells upon light irradiation.


Asunto(s)
Portadores de Fármacos/química , Nanopartículas/química , Fármacos Fotosensibilizantes/farmacología , Porfirinas/farmacología , Profármacos/farmacología , Adamantano/análogos & derivados , Adamantano/síntesis química , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Antineoplásicos/efectos de la radiación , Clorofilidas , Ciclodextrinas/síntesis química , Ciclodextrinas/farmacología , Ciclodextrinas/efectos de la radiación , Disulfuros/síntesis química , Disulfuros/farmacología , Disulfuros/efectos de la radiación , Portadores de Fármacos/síntesis química , Liberación de Fármacos , Humanos , Luz , Células MCF-7 , Fosforilcolina/análogos & derivados , Fosforilcolina/síntesis química , Fotoquimioterapia , Fármacos Fotosensibilizantes/síntesis química , Fármacos Fotosensibilizantes/efectos de la radiación , Ácidos Polimetacrílicos/síntesis química , Ácidos Polimetacrílicos/química , Porfirinas/síntesis química , Porfirinas/efectos de la radiación , Profármacos/síntesis química , Profármacos/efectos de la radiación , Especies Reactivas de Oxígeno/metabolismo
17.
Inorg Chem ; 57(22): 14409-14420, 2018 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-30365308

RESUMEN

A series of dinuclear octahedral PtIV complexes trans, trans, trans-[{Pt(N3)2(py)2(OH)(OC(O)CH2CH2C(O)NH)}2R] containing pyridine (py) and bridging dicarboxylate [R = -CH2CH2- (1), trans-1,2-C6H10- (2), p-C6H4- (3), -CH2CH2CH2CH2- (4)] ligands have been synthesized and characterized, including the X-ray crystal structures of complexes 1·2MeOH and 4, the first photoactivatable dinuclear PtIV complexes with azido ligands. The complexes are highly stable in the dark, but upon photoactivation with blue light (420 nm), they release the bridging ligand and mononuclear photoproducts. Upon irradiation with blue light (465 nm), they generate azidyl and hydroxyl radicals, detected using a 5,5-dimethyl-1-pyrroline N-oxide electron paramagnetic resonance spin trap, accompanied by the disappearance of the ligand-to-metal charge-transfer (N3 → Pt) band at ca. 300 nm. The dinuclear complexes are photocytotoxic to human cancer cells (465 nm, 4.8 mW/cm2, 1 h), including A2780 human ovarian and esophageal OE19 cells with IC50 values of 8.8-78.3 µM, whereas cisplatin is inactive under these conditions. Complexes 1, 3, and 4 are notably more photoactive toward cisplatin-resistant ovarian A2780cis compared to A2780 cells. Remarkably, all of the complexes were relatively nontoxic toward normal cells (MRC5 lung fibroblasts), with IC50 values >100 µM, even after irradiation. The introduction of an aromatic bridging ligand (3) significantly enhanced cellular uptake. The populations in the stages of the cell cycle remained unchanged upon treatment with complexes in the dark, while the population of the G2/M phase increased upon irradiation, suggesting that DNA is a target for these photoactivated dinuclear PtIV complexes. Liquid chromatography-mass spectrometry data show that the photodecomposition pathway of the dinuclear complexes results in the release of two molecules of mononuclear platinum(II) species. As a consequence, DNA binding of the dinuclear complexes after photoactivation in cell-free media is, in several respects, qualitatively similar to that of the photoactivated mononuclear complex FM-190. After photoactivation, they were 2-fold more effective in quenching the fluorescence of EtBr bound to DNA, forming DNA interstrand cross-links and unwinding DNA compared to the photoactivated FM-190.


Asunto(s)
Antineoplásicos/farmacología , Compuestos Organoplatinos/farmacología , Fármacos Fotosensibilizantes/farmacología , Profármacos/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/efectos de la radiación , Bovinos , Línea Celular Tumoral , ADN/química , ADN/metabolismo , Humanos , Ligandos , Luz , Compuestos Organoplatinos/síntesis química , Compuestos Organoplatinos/química , Compuestos Organoplatinos/efectos de la radiación , Fármacos Fotosensibilizantes/síntesis química , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/efectos de la radiación , Profármacos/síntesis química , Profármacos/química , Profármacos/efectos de la radiación , Estereoisomerismo
18.
J Am Chem Soc ; 140(30): 9721-9729, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29983046

RESUMEN

The delivery of controlled amounts of carbon monoxide (CO) to biological targets is of significant current interest. Very few CO-releasing compounds are currently known that can be rigorously controlled in terms of the location and amount of CO released. To address this deficiency, we report herein a new metal-free, visible-light-induced CO-releasing molecule (photoCORM) and its prodrug oxidized form, which offer new approaches to controlled, localized CO delivery. The new photoCORM, based on a 3-hydroxybenzo[ g]quinolone framework, releases 1 equiv of CO upon visible-light illumination under a variety of biologically relevant conditions. This nontoxic compound can be tracked prior to CO release using fluorescence microscopy and produces a nontoxic byproduct following CO release. An oxidized prodrug form of the photoCORM is reduced by cellular thiols, providing an approach toward activation in the reducing environment of cancer cells. Strong noncovalent affinity of the nonmetal photoCORM to albumin enables use of an albumin:photoCORM complex for targeted CO delivery to cancer cells. This approach produced cytotoxicity IC50 values among the lowest reported to date for CO delivery to cancer cells by a photoCORM. This albumin:photoCORM complex is also the first CO delivery system to produce significant anti-inflammatory effects when introduced at nanomolar photoCORM concentration.


Asunto(s)
Antiinflamatorios/farmacología , Antineoplásicos/farmacología , Profármacos/farmacología , Quinolonas/farmacología , Células A549 , Animales , Antiinflamatorios/metabolismo , Antiinflamatorios/efectos de la radiación , Antiinflamatorios/toxicidad , Antineoplásicos/metabolismo , Antineoplásicos/efectos de la radiación , Antineoplásicos/toxicidad , Monóxido de Carbono , Bovinos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Luz , Ratones , Profármacos/metabolismo , Profármacos/efectos de la radiación , Profármacos/toxicidad , Unión Proteica , Quinolonas/metabolismo , Quinolonas/efectos de la radiación , Quinolonas/toxicidad , Células RAW 264.7 , Albúmina Sérica Bovina/metabolismo
19.
J Inorg Biochem ; 179: 146-153, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29180165

RESUMEN

CHS-828 (N-(6-(4-chlorophenoxy)hexyl)-N'-cyano-N″-4-pyridyl guanidine) is an anticancer agent with low bioavailability and high systemic toxicity. Here we present an approach to improve the therapeutic profile of the drug using photolabile ruthenium complexes to generate light-activated prodrugs of CHS-828. Both prodrug complexes are stable in the dark but release CHS-828 when irradiated with visible light. The complexes are water-soluble and accumulate in tumour cells in very high concentrations, predominantly in the mitochondria. Both prodrug complexes are significantly less cyototoxic than free CHS-828 in the dark but their toxicity increases up to 10-fold in combination with visible light. The cellular responses to light treatment are consistent with release of the cytotoxic CHS-828 ligand.


Asunto(s)
Antineoplásicos/farmacología , Complejos de Coordinación/farmacología , Cianuros/farmacología , Guanidinas/farmacología , Profármacos/farmacología , Rutenio/química , Células A549 , Antineoplásicos/síntesis química , Antineoplásicos/efectos de la radiación , Complejos de Coordinación/síntesis química , Complejos de Coordinación/efectos de la radiación , Cianuros/efectos de la radiación , ADN/química , Guanidinas/efectos de la radiación , Humanos , Concentración 50 Inhibidora , Luz , Células MCF-7 , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Profármacos/síntesis química , Profármacos/efectos de la radiación , Especies Reactivas de Oxígeno/metabolismo
20.
J Med Chem ; 60(21): 8923-8932, 2017 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-28991460

RESUMEN

A unique, dual-function, photoactivatable anticancer prodrug, RuEuL, has been tailored that features a ruthenium(II) complex linked to a cyclen-europium chelate via a π-conjugated bridge. Under irradiation at 488 nm, the dark-inactive prodrug undergoes photodissociation, releasing the DNA-damaging ruthenium species. Under evaluation-window irradiation (λirr = one-photon 350 nm or two-photon 700 nm), the drug delivery process can be quantitatively monitored in real-time because of the long-lived red europium emission. Linear relationships between released drug concentration and ESI-MS or luminescence responses are established. Finally, the efficiency of the new prodrug is demonstrated both in vitro RuEuL anticancer prodrug over some existing ones and open the way for decisive improvements in multipurpose prodrugs.


Asunto(s)
Antineoplásicos/química , Europio/química , Profármacos/química , Rutenio/química , Animales , Liberación de Fármacos/efectos de la radiación , Monitoreo de Drogas/métodos , Humanos , Luz , Fotólisis , Profármacos/efectos de la radiación , Análisis Espectral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...