Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros











Intervalo de año de publicación
1.
Inorg Chem ; 63(17): 7973-7983, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38616353

RESUMEN

Dysregulated cathepsin activity is linked to various human diseases including metabolic disorders, autoimmune conditions, and cancer. Given the overexpression of cathepsin in the tumor microenvironment, cathepsin inhibitors are promising pharmacological agents and drug delivery vehicles for cancer treatment. In this study, we describe the synthesis and photochemical and biological assessment of a dual-action agent based on ruthenium that is conjugated with a cathepsin inhibitor, designed for both photodynamic therapy (PDT) and photochemotherapy (PCT). The ruthenium-cathepsin inhibitor conjugate was synthesized through an oxime click reaction, combining a pan-cathepsin inhibitor based on E64d with the Ru(II) PCT/PDT fragment [Ru(dqpy)(dppn)], where dqpy = 2,6-di(quinoline-2-yl)pyridine and dppn = benzo[i]dipyrido[3,2-a:2',3'-c]phenazine. Photochemical investigations validated the conjugate's ability to release a triazole-containing cathepsin inhibitor for PCT and to generate singlet oxygen for PDT upon exposure to green light. Inhibition studies demonstrated the conjugate's potent and irreversible inactivation of purified and intracellular cysteine cathepsins. Two Ru(II) PCT/PDT agents based on the [Ru(dqpy)(dppn)] moiety were evaluated for photoinduced cytotoxicity in 4T1 murine triple-negative breast cancer cells, L929 fibroblasts, and M0, M1, and M2 macrophages. The cathepsin inhibitor conjugate displayed notable selectivity for inducing cell death under irradiation compared to dark conditions, mitigating toxicity in the dark observed with the triazole control complex [Ru(dqpy)(dppn)(MeTz)]2+ (MeTz = 1-methyl-1H-1,2,4-triazole). Notably, our lead complex is among a limited number of dual PCT/PDT agents activated with green light.


Asunto(s)
Catepsinas , Luz , Fotoquimioterapia , Fármacos Fotosensibilizantes , Rutenio , Humanos , Rutenio/química , Rutenio/farmacología , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/síntesis química , Catepsinas/antagonistas & inhibidores , Catepsinas/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Estructura Molecular , Línea Celular Tumoral , Complejos de Coordinación/farmacología , Complejos de Coordinación/química , Complejos de Coordinación/síntesis química , Animales , Ratones , Supervivencia Celular/efectos de los fármacos , Luz Verde
2.
Chem Sci ; 13(7): 1933-1945, 2022 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-35308843

RESUMEN

Two new tris-heteroleptic Ru(ii) complexes with triphenylphosphine (PPh3) coordination, cis-[Ru(phen)2(PPh3)(CH3CN)]2+ (1a, phen = 1,10-phenanthroline) and cis-[Ru(biq)(phen)(PPh3)(CH3CN)]2+ (2a, biq = 2,2'-biquinoline), were synthesized and characterized for photochemotherapeutic applications. Upon absorption of visible light, 1a exchanges a CH3CN ligand for a solvent water molecule. Surprisingly, the steady-state irradiation of 2a followed by electronic absorption and NMR spectroscopies reveals the photosubstitution of the PPh3 ligand. Phosphine photoinduced ligand exchange with visible light from a Ru(ii) polypyridyl complex has not previously been reported, and calculations reveal that it results from a trans-type influence in the excited state. Complexes 1a and 2a are not toxic against the triple negative breast cancer cell line MDA-MB-231 in the dark, but upon irradiation with blue light, the activity of both complexes increases by factors of >4.2 and 5.8, respectively. Experiments with PPh3 alone show that the phototoxicity observed for 2a does not arise from the released phosphine ligand, indicating the role of the photochemically generated ruthenium aqua complex on the biological activity. These complexes represent a new design motif for the selective release of PPh3 and CH3CN for use in photochemotherapy.

3.
Chemistry ; 28(24): e202104430, 2022 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-35235227

RESUMEN

Tumor associated macrophages (TAMs) suppress the cancer immune response and are a key target for immunotherapy. The effects of ruthenium and rhodium complexes on TAMs have not been well characterized. To address this gap in the field, a panel of 22 dirhodium and ruthenium complexes were screened against three subtypes of macrophages, triple-negative breast cancer and normal breast tissue cells. Experiments were carried out in 2D and biomimetic 3D co-culture experiments with and without irradiation with blue light. Leads were identified with cell-type-specific toxicity toward macrophage subtypes, cancer cells, or both. Experiments with 3D spheroids revealed complexes that sensitized the tumor models to the chemotherapeutic doxorubicin. Cell surface exposure of calreticulin, a known facilitator of immunogenic cell death (ICD), was increased upon treatment, along with a concomitant reduction in the M2-subtype classifier arginase. Our findings lay a strong foundation for the future development of ruthenium- and rhodium-based chemotherapies targeting TAMs.


Asunto(s)
Rodio , Rutenio , Neoplasias de la Mama Triple Negativas , Línea Celular Tumoral , Humanos , Inmunoterapia , Rodio/farmacología , Rutenio/farmacología , Macrófagos Asociados a Tumores
4.
Photochem Photobiol ; 98(2): 378-388, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34866185

RESUMEN

We report the synthesis, photochemical and biological characterization of two new Ru(II) photoactivated complexes based on [Ru(tpy)(Me2 bpy)(L)]2+ (tpy = 2,2':6',2''-terpyridine, Me2 bpy = 6,6'-dimethyl-2,2'-bipyridine), where L = pyridyl-BODIPY (pyBOD). Two pyBOD ligands were prepared bearing flanking hydrogen or iodine atoms. Ru(II)-bound BODIPY dyes show a red-shift of absorption maxima relative to the free dyes and undergo photodissociation of BODIPY ligands with green light irradiation. Addition of iodine into the BODIPY ligand facilitates intersystem crossing, which leads to efficient singlet oxygen production in the free dye, but also enhances quantum yield of release of the BODIPY ligand from Ru(II). This represents the first report of a strategy to enhance photodissociation quantum yields through the heavy-atom effect in Ru(II) complexes. Furthermore, Ru(II)-bound BODIPY dyes display fluorescence turn-on once released, with a lead analog showing nanomolar EC50 values against triple negative breast cancer cells, >100-fold phototherapeutic indexes under green light irradiation, and higher selectivity toward cancer cells as compared to normal cells than the corresponding free BODIPY photosensitizer. Conventional Ru(II) photoactivated complexes require nonbiorthogonal blue light for activation and rarely show submicromolar potency to achieve cell death. Our study represents an avenue for the improved photochemistry and potency of future Ru(II) complexes.


Asunto(s)
Yodo , Rutenio , Colorantes , Ligandos , Fármacos Fotosensibilizantes/química , Rutenio/química
5.
Inorg Chem ; 60(24): 18964-18974, 2021 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-34846875

RESUMEN

The synthesis, chemical and biological characterization of seven Ru(II) polypyridyl complexes containing acetylacetonate (acac) ligands are reported. Electronic absorption spectra were determined and electrochemical potentials consistent with Ru(III/II) couples ranging from +0.60 to +0.73 V vs Ag/AgCl were measured. A series of complexes were screened against MDA-MB-231, DU-145, and MCF-10A cell lines to evaluate their cytotoxicities in cancer and normal cell lines. Although most complexes were either nontoxic or equipotent in cancer cells and normal cell lines, compound 1, [Ru(dpqy)(acac)(py)](PF6), where dqpy is 2,6-di(quinolin-2-yl)pyridine, showed up to 2.5:1.0 selectivity for cancer as compared to normal cells, along with nanomolar EC50 values in MDA-MB-231 cells. Lipophilicity, determined as the octanol/water partition coefficient, log Po/w, ranged from -0.33 (0.06) to 1.15 (0.10) for the complexes. Although cytotoxicity was not correlated with electrochemical potentials, a moderate linear correlation between lipophilicity and toxicities was observed. Cell death mechanism studies indicated that several of the Ru-acac compounds, including 1, induce apoptosis in MDA-MB-231 cells.


Asunto(s)
Apoptosis
6.
Chem Sci ; 12(36): 12056-12067, 2021 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-34667571

RESUMEN

A series of five ruthenium complexes containing triphenyl phosphine groups known to enhance both cellular penetration and photoinduced ligand exchange, cis-[Ru(bpy)2(P(p-R-Ph)3)(CH3CN)]2+, where bpy = 2,2'-bipyridine and P(p-R-Ph)3 represent para-substituted triphenylphosphine ligands with R = -OCH3 (1), -CH3 (2) -H (3), -F (4), and -CF3 (5), were synthesized and characterized. The photolysis of 1-5 in water with visible light (λ irr ≥ 395 nm) results in the substitution of the coordinated acetonitrile with a solvent molecule, generating the corresponding aqua complex as the single photoproduct. A 3-fold variation in quantum yield was measured with 400 nm irradiation, Φ 400, where 1 is the most efficient with a Φ 400 = 0.076(2), and 5 the least photoactive complex, with Φ 400 = 0.026(2). This trend is unexpected based on the red-shifted metal-to-ligand charge transfer (MLCT) absorption of 1 as compared to that of 5, but can be correlated to the substituent Hammett para parameters and pK a values of the ancillary phosphine ligands. Complexes 1-5 are not toxic towards the triple negative breast cancer cell line MDA-MB-231 in the dark, but 3 and 5 are >4.2 and >19-fold more cytotoxic upon irradiation with blue light, respectively. A number of experiments point to apoptosis, and not to necrosis or necroptosis, as the mechanism of cell death by 5 upon irradiation. These findings provide a foundation for understanding the role of phosphine ligands on photoinduced ligand substitution and show the enhancement afforded by -CF3 groups on photochemotherapy, which will aid the future design of photocages for photochemotherapeutic drug delivery.

7.
J Am Chem Soc ; 143(24): 9191-9205, 2021 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-34110801

RESUMEN

We report the synthesis and photochemical and biological characterization of the first selective and potent metal-based inhibitors of cytochrome P450 3A4 (CYP3A4), the major human drug metabolizing enzyme. Five Ru(II)-based derivatives were prepared from two analogs of the CYP3A4 inhibitor ritonavir, 4 and 6: [Ru(tpy)(L)(6)]Cl2 (tpy = 2,2':6',2″-terpyridine) with L = 6,6'-dimethyl-2,2'-bipyridine (Me2bpy; 8), dimethylbenzo[i]dipyrido[3,2-a:2',3'-c]phenazine (Me2dppn; 10) and 3,6-dimethyl-10,15-diphenylbenzo[i]dipyrido[3,2-a:2',3'-c]phenazine (Me2Ph2dppn; 11), [Ru(tpy)(Me2bpy)(4)]Cl2 (7) and [Ru(tpy)(Me2dppn)(4)]Cl2 (9). Photochemical release of 4 or 6 from 7-11 was demonstrated, and the spectrophotometric evaluation of 7 showed that it behaves similarly to free 4 (type II heme ligation) after irradiation with visible light but not in the dark. Unexpectedly, the intact Ru(II) complexes 7 and 8 were found to inhibit CYP3A4 potently and specifically through direct binding to the active site without heme ligation. Caged inhibitors 9-11 showed dual action properties by combining photoactivated dissociation of 4 or 6 with efficient 1O2 production. In prostate adenocarcinoma DU-145 cells, compound 9 had the best synergistic effect with vinblastine, the anticancer drug primarily metabolized by CYP3A4 in vivo. Thus, our study establishes a new paradigm in CYP inhibition using metalated complexes and suggests possible utilization of photoactive CYP3A4 inhibitory compounds in clinical applications, such as enhancement of therapeutic efficacy of anticancer drugs.


Asunto(s)
Antineoplásicos/farmacología , Complejos de Coordinación/farmacología , Inhibidores del Citocromo P-450 CYP3A/farmacología , Citocromo P-450 CYP3A/metabolismo , Fármacos Fotosensibilizantes/farmacología , Rutenio/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Complejos de Coordinación/síntesis química , Complejos de Coordinación/química , Inhibidores del Citocromo P-450 CYP3A/síntesis química , Inhibidores del Citocromo P-450 CYP3A/química , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Modelos Moleculares , Estructura Molecular , Fármacos Fotosensibilizantes/síntesis química , Fármacos Fotosensibilizantes/química , Rutenio/química
8.
Inorg Chem ; 59(6): 3919-3933, 2020 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-32096986

RESUMEN

We report the synthesis and photochemical and biological characterization of Ru(II) complexes containing π-expansive ligands derived from dimethylbenzo[i]dipyrido[3,2-a:2',3'-c]phenazine (Me2dppn) adorned with flanking aryl substituents. Late-stage Suzuki couplings produced Me2dppn ligands substituted at the 10 and 15 positions with phenyl (5), 2,4-dimethylphenyl (6), and 2,4-dimethoxyphenyl (7) groups. Complexes of the general formula [Ru(tpy)(L)(py)](PF6)2 (8-10), where L = 4-7, were characterized and shown to have dual photochemotherapeutic (PCT) and photodynamic therapy (PDT) behavior. Quantum yields for photodissociation of monodentate pyridines from 8-10 were about 3 times higher than that of parent complex [Ru(tpy)(Me2dppn)(py)](PF6)2 (1), whereas quantum yields for singlet oxygen (1O2) production were ∼10% lower than that of 1. Transient absorption spectroscopy indicates that 8-10 possess long excited state lifetimes (τ = 46-50 µs), consistent with efficient 1O2 production through population and subsequent decay of ligand-centered 3ππ* excited states. Complexes 8-10 displayed greater lipophilicity relative to 1 and association to DNA but do not intercalate between the duplex base pairs. Complexes 1 and 8-10 showed photoactivated toxicity in breast and prostate cancer cell lines with phototherapeutic indexes, PIs, as high as >56, where the majority of cell death was achieved 4 h after treatment with Ru(II) complexes and light. Flow cytometric data and rescue experiments were consistent with necrotic cell death mediated by the production of reactive oxygen species, especially 1O2. Collectively, this study confirms that DNA intercalation by Ru(II) complexes with π-expansive ligands is not required to achieve photoactivated cell death.


Asunto(s)
Antineoplásicos/farmacología , Complejos de Coordinación/farmacología , Fármacos Fotosensibilizantes/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Antineoplásicos/efectos de la radiación , Línea Celular Tumoral , Complejos de Coordinación/síntesis química , Complejos de Coordinación/metabolismo , Complejos de Coordinación/efectos de la radiación , ADN/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Radical Hidroxilo/metabolismo , Ligandos , Luz , Necrosis/inducido químicamente , Fármacos Fotosensibilizantes/síntesis química , Fármacos Fotosensibilizantes/metabolismo , Fármacos Fotosensibilizantes/efectos de la radiación , Rutenio/química , Oxígeno Singlete/metabolismo
9.
ACS Chem Biol ; 14(12): 2833-2840, 2019 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-31750642

RESUMEN

Acquired resistance to apoptotic agents is a long-standing challenge in cancer treatment. Cathepsin B (CTSB) is an enzyme which, among many essential functions, promotes apoptosis during cellular stress through regulation of intracellular proteolytic networks on the minute time scale. Recent data indicate that CTSB inhibition may be a promising method to steer cells away from apoptotic death toward necrosis, a mechanism of cell death that can overcome resistance to apoptotic agents, stimulate an immune response and promote antitumor immunity. Unfortunately, rapid and selective intracellular inactivation of CTSB has not been possible. However, here we report on the synthesis and characterization of photochemical and biological properties of BODIPY-caged inhibitors of CTSB that are cell permeable, highly selective and activated rapidly upon exposure to visible light. Intriguingly, these compounds display tunable photophysical and biological properties based on substituents bound directly to boron. Me2BODIPY-caged compound 8 displays the dual-action capability of light-accelerated CTSB inhibition and singlet oxygen production from a singular molecular entity. The dual-action capacity of 8 leads to a rapid necrotic response in MDA-MB-231 triple negative breast cancer cells with high phototherapeutic indexes (>30) and selectivity vs noncancerous cells that neither CTSB inhibition nor photosensitization gives alone. Our work confirms that singlet oxygen production and CTSB inactivation is highly synergistic and a promising method for killing cancer cells. Furthermore, this ability to trigger intracellular inactivation of CTSB with light provides researchers with a powerful photochemical tool for probing biochemical processes on short time scales.


Asunto(s)
Apoptosis/efectos de los fármacos , Compuestos de Boro/química , Catepsina B/antagonistas & inhibidores , Inhibidores de Cisteína Proteinasa/farmacología , Luz , Neoplasias/patología , Línea Celular Tumoral , Inhibidores de Cisteína Proteinasa/química , Humanos , Estrés Oxidativo
10.
J Am Chem Soc ; 140(43): 14367-14380, 2018 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-30278123

RESUMEN

Dual action agents containing a cysteine protease inhibitor and Ru-based photosensitizer for photodynamic therapy (PDT) were designed, synthesized, and validated in 2D culture and 3D functional imaging assays of triple-negative human breast cancer (TNBC). These combination agents deliver and release Ru-based PDT agents to tumor cells and cause cancer cell death upon irradiation with visible light, while at the same time inactivating cathespin B (CTSB), a cysteine protease strongly associated with invasive and metastatic behavior. In total five Ru-based complexes were synthesized with the formula [Ru(bpy)2(1)](O2CCF3)2 (3), where bpy = 2,2'-bipyridine and 1 = a bipyridine-based epoxysuccinyl inhibitor; [Ru(tpy)(NN)(2)](PF6)2, where tpy = terpiridine, 2 = a pyridine-based epoxysuccinyl inhibitor and NN = 2,2'-bipyridine (4); 6,6'-dimethyl-2,2'-bipyridine (5); benzo[ i]dipyrido[3,2- a:2',3'- c]phenazine (6); and 3,6-dimethylbenzo[ i]dipyrido[3,2- a:2',3'- c]phenazine (7). Compound 3 contains a [Ru(bpy)3]2+ fluorophore and was designed to track the subcellular localization of the conjugates, whereas compounds 4-7 were designed to undergo either photoactivated ligand dissociation and/or singlet oxygen generation. Photochemical studies confirmed that complexes 5 and 7 undergo photoactivated ligand dissociation, whereas 6 and 7 generate singlet oxygen. Inhibitors 1-7 all potently and irreversibly inhibit CTSB. Compounds 4-7 were evaluated against MDA-MB-231 TNBC and MCF-10A breast epithelial cells in 2D and 3D culture for effects on proteolysis and cell viability under dark and light conditions. Collectively, these data reveal that 4-7 potently inhibit dye-quenched (DQ) collagen degradation, whereas only compound 7 causes efficient cell death under light conditions, consistent with its ability to release a Ru(II)-based photosensitizer and to also generate 1O2.


Asunto(s)
Antineoplásicos/farmacología , Complejos de Coordinación/farmacología , Péptido Hidrolasas/metabolismo , Fármacos Fotosensibilizantes/farmacología , Inhibidores de Proteasas/farmacología , Rutenio/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Complejos de Coordinación/síntesis química , Complejos de Coordinación/química , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Cinética , Fármacos Fotosensibilizantes/síntesis química , Fármacos Fotosensibilizantes/química , Inhibidores de Proteasas/síntesis química , Inhibidores de Proteasas/química , Rutenio/química , Termodinámica
11.
Inorg Chem ; 57(13): 7881-7891, 2018 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-29882662

RESUMEN

Two new Re(I)- and Ru(II)-based inhibitors were synthesized with the formulas [Re(phen)(CO)3(1)](OTf) (7; phen = 1,10-phenanthroline, OTf = trifluoromethanesulfonate) and [Ru(bpy)2(2)](Cl)2 (8; bpy = 2,2'-bipyridine), where 1 and 2 are the analogues of CLIK-148, an epoxysuccinyl-based cysteine cathepsin L inhibitor (CTSL). Compounds 7 and 8 were characterized using various spectroscopic techniques and elemental analysis; 7 and 8 both show exceptionally long excited state lifetimes. Re(I)-based complex 7 inhibits CTSL in the low nanomolar range, affording a greater than 16-fold enhancement of potency relative to the free inhibitor 1 with a second-order rate constant of 211000 ± 42000 M-1 s-1. Irreversible ligation of 7 to papain, a model of CTSL, was analyzed with mass spectroscopy, and the major peak shown at 24283 au corresponds to that of papain-1-Re(CO)3(phen). Compound 7 was well tolerated by DU-145 prostate cancer cells, with toxicity evident only at high concentrations. Treatment of DU-145 cells with 7 followed by imaging via confocal microscopy showed substantial intracellular fluorescence that can be blocked by the known CTSL inhibitor CLIK-148, consistent with the ability of 7 to label CTSL in living cells. Overall this study reveals that a Re(I) complex can be attached to an enzyme inhibitor and enhance potency and selectivity for a medicinally important target, while at the same time allowing new avenues for tracking and quantification due to long excited state lifetimes and non-native element composition.


Asunto(s)
Catepsina L/antagonistas & inhibidores , Compuestos Organometálicos/química , Compuestos Organometálicos/farmacología , Renio/química , Rutenio/química , Catepsina L/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Sustancias Luminiscentes/química , Sustancias Luminiscentes/farmacología , Modelos Moleculares , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , Conformación Proteica
12.
Acc Chem Res ; 51(6): 1415-1421, 2018 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-29870227

RESUMEN

Metal complexes have many proven applications in the caging and photochemical release of biologically active compounds. Photocaging groups derived from Ru(II) traditionally have been composed of ancillary ligands that are planar and bi- or tridentate, such as 2,2'-bipyridine (bpy), 2,2':6',2″-terpyridine (tpy), and 1,10-phenanthroline (phen). Complexes bearing ancillary ligands with denticities higher than three represent a new class of Ru(II)-based photocaging groups that are grossly underdeveloped. Because high-denticity ancillary ligands provide the ability to increase the structural rigidity and control the stereochemistry, our groups initiated a research program to explore the applications of such ligands in Ru(II)-based photocaging. Ru(TPA), bearing the tetradentate ancillary ligand tris(2-pyridylmethyl)amine (TPA), has been successfully utilized to effectively cage nitriles and aromatic heterocycles. Nitriles and aromatic heterocycles caged by the Ru(TPA) group show excellent stability in aqueous solutions in the dark, and the complexes can selectively release the caged molecules upon irradiation with light. Ru(TPA) is applicable as a photochemical agent to offer precise spatiotemporal control over biological activity without undesired toxicity. In addition, Ru(II) polypyridyl complexes with desired photochemical properties can be synthesized and identified by solid-phase synthesis, and the resulting complexes show properties to similar to those of complexes obtained by solution-phase synthesis. Density functional theory (DFT) calculations reveal that orbital mixing between the π* orbitals of the ancillary ligand and the Ru-N dσ* orbital is essential for ligand photodissociation in these complexes. Furthermore, the introduction of steric bulk enhances the photoliability of the caged molecules, validating that steric effects can largely influence the quantum efficiency of photoinduced ligand exchange in Ru(II) polypyridyl complexes. Recently, two new photocaging groups, Ru(cyTPA) and Ru(1-isocyTPQA), have been designed and synthesized for caging of nitriles and aromatic heterocycles, and these complexes exhibit unique photochemical properties distinct from those derived from Ru(TPA). Notably, the unusually greater quantum efficiency for the ligand exchange in [Ru(1-isocyTPQA)(MeCN)2](PF6)2, Φ400 = 0.033(3), uncovers a trans-type effect in the triplet metal-to-ligand charge transfer (3MLCT) state that enhances photoinduced ligand exchange in a new manner. DFT calculations and ultrafast transient spectroscopy reveal that the lowest-energy triplet state in [Ru(1-isocyTPQA)(MeCN)2](PF6)2 is a highly mixed 3MLCT/3ππ* excited state rather than a triplet metal-centered ligand-field (3LF) excited state; the latter is generally accepted for ligand photodissociation. In addition, Mulliken spin density calculations indicate that a majority of the spin density in [Ru(1-isocyTPQA)(MeCN)2](PF6)2 is localized on the isoquinoline arm, which is opposite to the cis MeCN, rather than on the ruthenium center. This significantly weakens the Ru-N6 ( cis MeCN) bond, which then promotes the ligand photodissociation. This newly discovered effect gives a clearer perception of the interplay between the 3MLCT and 3LF excited states of Ru(II) polypyridyl complexes, which may be useful in the design and applications of ruthenium complexes in the areas of photoactivated drug delivery and photosensitizers.


Asunto(s)
Complejos de Coordinación/química , Piridinas/química , Rutenio/química , Complejos de Coordinación/síntesis química , Complejos de Coordinación/efectos de la radiación , Teoría Funcional de la Densidad , Ligandos , Modelos Químicos , Nitrilos/síntesis química , Nitrilos/química , Nitrilos/efectos de la radiación , Piridinas/síntesis química , Piridinas/efectos de la radiación
13.
Chem Commun (Camb) ; 54(41): 5193-5196, 2018 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-29707728

RESUMEN

Ru(ii)-polypyridyl cages with sterically bulky bidentate ligands provide efficient photochemical release of the anticancer drug imatinib using low energy visible light, imparting spatiotemporal control over drug bioavailability. The light-activated drug release is maintained when the Ru(ii) cage is covalently coupled to an antibody, which is expected to localize selectively on the tumor.


Asunto(s)
Anticuerpos/química , Complejos de Coordinación/química , Mesilato de Imatinib/química , Luz , Procesos Fotoquímicos , Piridinas/química , Rutenio/química , Estructura Molecular
14.
Dalton Trans ; 47(34): 11851-11858, 2018 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-29741184

RESUMEN

A new complex, [Ru(tpy)(dppn)(Cbz-Leu-NHCH2CN)]2+ (1, tpy = 2,2':6',2''-terpyridine, dppn = benzo[i]dipyrido[3,2-a:2',3'-c]phenazine) was synthesized and its photochemical properties were investigated. This complex undergoes photorelease of the Cbz-Leu-NHCH2CN ligand, a known cathepsin K inhibitor, with a quantum yield, Φ450, of 0.0012(4) in water (λirr = 450 nm). In addition, 1 sensitizes the production of singlet oxygen upon visible light irradiation with quantum yield, ΦΔ, of 0.64(3) in CH3OH. The photophysical properties of 1 were compared with those of [Ru(tpy)(bpy)(Cbz-Leu-NHCH2CN)]2+ (2, bpy = 2,2'-bipyridine), [Ru(tpy)(dppn)(CH3CN)]2+ (3), and [Ru(tpy)(bpy)(CH3CN)]2+ (4) to evaluate the effect of the release of the Cbz-Leu-NHCH2CN inhibitor relative to the CH3CN ligand, as well as the role of dppn as the bidentate ligand for 1O2 production instead of bpy. Nanosecond transient absorption spectroscopy confirms the formation of the long-lived dppn-centered 3ππ* state in 1 and 3 with a maximum at ∼540 nm and τ ∼20 µs in deaerated acetonitrile. Complexes 1 and 3 are able to cause photoinduced damage to DNA (λirr ≥ 395 nm), whereas 2 and 4 do not photocleave DNA under similar experimental conditions. These results suggest that 1 is a promising agent for dual activity, both releasing a drug and producing singlet oxygen, and is poised to exhibit enhanced biological activity in phototochemotherapy upon irradiation with visible light.


Asunto(s)
Catepsina K/metabolismo , Complejos de Coordinación/farmacología , Inhibidores Enzimáticos/farmacología , Fotoquimioterapia , Rutenio/química , Oxígeno Singlete/metabolismo , Acetonitrilos/química , Complejos de Coordinación/síntesis química , Complejos de Coordinación/química , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Ligandos , Metanol/química , Neoplasias/tratamiento farmacológico , Fenazinas/química , Fármacos Fotosensibilizantes/síntesis química , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología , Plásmidos/metabolismo , Piridinas/química , Agua/química
15.
Chem Commun (Camb) ; 54(11): 1280-1290, 2018 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-29323683

RESUMEN

Photocaging allows for precise spatiotemporal control over the release of biologically active compounds with light. Most photocaged molecules employ organic photolabile protecting groups; however, biologically active compounds often contain functionalities such as nitriles and aromatic heterocycles that cannot be caged with organic groups. Despite their prevalence, only a few studies have reported successful caging of nitriles and aromatic heterocycles. Recently, Ru(ii)-based photocaging has emerged as a powerful method for the release of bioactive molecules containing these functional groups, in many cases providing high levels of spatial and temporal control over biological activity. This Feature Article discusses recent developments in applying Ru(ii)-based photocaging towards biological problems. Our groups designed and synthesized Ru(ii)-based platforms for the photoinduced delivery of cysteine protease and cytochrome P450 inhibitors in order to achieve selective control over enzyme inhibition. We also reported Ru(ii) photocaging groups derived from higher-denticity ancillary ligands that possess photophysical and photochemical properties distinct from more traditional Ru(ii)-based caging groups. In addition, for the first time, we are able to rapidly synthesize and screen Ru(ii) polypyridyl complexes that elicit desired properties by solid-phase synthesis. Finally, our work also defined steric and orbital mixing effects that are important factors in controlling photoinduced ligand exchange.


Asunto(s)
Complejos de Coordinación/efectos de la radiación , Liberación de Fármacos , Nitrilos/efectos de la radiación , Piridinas/efectos de la radiación , Rutenio/química , Catepsinas/antagonistas & inhibidores , Línea Celular Tumoral , Complejos de Coordinación/síntesis química , Complejos de Coordinación/química , Inhibidores de Cisteína Proteinasa/química , Inhibidores de Cisteína Proteinasa/efectos de la radiación , Inhibidores Enzimáticos del Citocromo P-450/química , Inhibidores Enzimáticos del Citocromo P-450/efectos de la radiación , Humanos , Luz , Estructura Molecular , Nitrilos/química , Piridinas/síntesis química , Piridinas/química , Esteroide 17-alfa-Hidroxilasa/química
16.
Chem Commun (Camb) ; 53(26): 3673-3676, 2017 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-28304025
17.
Biol Chem ; 397(6): 571-82, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-26901495

RESUMEN

Collagenolytic activity of cathepsin K is important for many physiological and pathological processes including osteoclast-mediated bone degradation, macrophage function and fibroblast-mediated matrix remodeling. Here, we report application of a light-activated inhibitor for controlling activity of cathepsin K in a 3D functional imaging assay. Using prostate carcinoma cell line engineered to overexpress cathepsin K, we demonstrate the utility of the proteolytic assay in living tumor spheroids for the evaluation and quantification of the inhibitor effects on cathepsin K-mediated collagen I degradation. Importantly, we also show that utilizing the ruthenium-caged version of a potent nitrile cathepsin K inhibitor (4), cis-[Ru(bpy)2(4)2](BF4)2 (5), offers significant advantage in terms of effective concentration of the inhibitor and especially its light-activated control in the 3D assay. Our results suggest that light activation provides a suitable, attractive approach for spatial and temporal control of proteolytic activity, which remains a critical, unmet need in treatment of human diseases, especially cancer.


Asunto(s)
Catepsina K/antagonistas & inhibidores , Imagenología Tridimensional , Luz , Neoplasias de la Próstata/patología , Inhibidores de Proteasas/farmacología , Catepsina K/genética , Línea Celular Tumoral , Colágeno Tipo I/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Masculino , Neoplasias de la Próstata/genética , Proteolisis/efectos de los fármacos , Proteolisis/efectos de la radiación
18.
PLoS One ; 10(11): e0142527, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26562785

RESUMEN

The cysteine protease cathepsin B has been causally linked to progression and metastasis of breast cancers. We demonstrate inhibition by a dipeptidyl nitrile inhibitor (compound 1) of cathepsin B activity and also of pericellular degradation of dye-quenched collagen IV by living breast cancer cells. To image, localize and quantify collagen IV degradation in real-time we used 3D pathomimetic breast cancer models designed to mimic the in vivo microenvironment of breast cancers. We further report the synthesis and characterization of a caged version of compound 1, [Ru(bpy)2(1)2](BF4)2 (compound 2), which can be photoactivated with visible light. Upon light activation, compound 2, like compound 1, inhibited cathepsin B activity and pericellular collagen IV degradation by the 3D pathomimetic models of living breast cancer cells, without causing toxicity. We suggest that caged inhibitor 2 is a prototype for cathepsin B inhibitors that can control both the site and timing of inhibition in cancer.


Asunto(s)
Catepsina B/antagonistas & inhibidores , Inhibidores de Cisteína Proteinasa/farmacología , Compuestos de Rutenio/farmacología , Microambiente Tumoral/efectos de los fármacos , Biocatálisis/efectos de los fármacos , Catepsina B/metabolismo , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Colágeno Tipo IV/metabolismo , Inhibidores de Cisteína Proteinasa/química , Diagnóstico por Imagen/métodos , Relación Dosis-Respuesta a Droga , Humanos , Luz , Microscopía Confocal , Estructura Molecular , Procesos Fotoquímicos/efectos de la radiación , Proteolisis/efectos de los fármacos , Compuestos de Rutenio/química , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
19.
Inorg Chem ; 54(16): 8003-11, 2015 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-26244447

RESUMEN

Metal complexes that release ligands upon photoexcitation are important tools for biological research and show great potential as highly specific therapeutics. Upon excitation with visible light, [Ru(TQA)(MeCN)2](2+) [TQA = tris(2-quinolinylmethyl)amine] exchanges one of the two acetonitriles (MeCNs), whereas [Ru(DPAbpy)MeCN](2+) [DPAbpy = N-(2,2'-bipyridin-6-yl)-N,N-bis(pyridin-2-ylmethyl)amine] does not release MeCN. Furthermore, [Ru(TQA)(MeCN)2](2+) is highly selective for release of the MeCN that is perpendicular to the plane of the two axial quinolines. Density functional theory calculations provide a clear explanation for the photodissociation behavior of these two complexes. Excitation by visible light and intersystem crossing leads to a six-coordinate (3)MLCT state. Dissociation of acetonitrile can occur after internal conversion to a dissociative (3)MC state, which has an occupied dσ* orbital that interacts in an antibonding fashion with acetonitrile. For [Ru(TQA)(MeCN)2](2+), the dissociative (3)MC state is lower than the (3)MLCT state. In contrast, the (3)MC state of [Ru(DPAbpy)MeCN](2+) that releases acetonitrile has an energy higher than that of the (3)MLCT state, indicating dissociation is unfavorable. These results are consistent with the experimental observations that efficient photodissociation of acetonitrile occurs for [Ru(TQA)(MeCN)2](2+) but not for [Ru(DPAbpy)MeCN](2+). For the release of the MeCN ligand in [Ru(TQA)(MeCN)2](2+) that is perpendicular to the axial quinoline rings, the (3)MLCT state has an occupied quinoline π* orbital that can interact with a dσ* Ru-NCCH3 antibonding orbital as the Ru-NCCH3 bond is stretched and the quinolines bend toward the departing acetonitrile. This reduces the barrier for the formation of the dissociative (3)MC state, leading to the selective photodissociation of this acetonitrile. By contrast, when the acetonitrile is in the plane of the quinolines or bpy, no interaction occurs between the ligand π* orbital and the dσ* Ru-NCCH3 orbital, resulting in high barriers for conversion to the corresponding (3)MC structures and no release of acetonitrile.


Asunto(s)
Acetonitrilos/química , Compuestos Organometálicos/química , Procesos Fotoquímicos , Piridinas/química , Teoría Cuántica , Rutenio/química , Ligandos , Modelos Moleculares , Conformación Molecular , Propiedades de Superficie
20.
J Cell Biochem ; 113(8): 2567-75, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22415943

RESUMEN

X-linked inhibitor of apoptosis protein (XIAP), inhibits the initiation and execution phases of the apoptotic pathway. XIAP is the most potent member of the inhibitor of apoptosis protein (IAP) family of the endogenous caspase inhibitors. Therefore, targeting XIAP may be a promising strategy for the treatment of apoptosis-resistant malignancies. In this study, we systematically studied the relationships of chemical structures of several novel ligands to their zinc (Zn)-binding ability, molecular target XIAP, and tumor cell death-inducing activity. We show that treatment of PC-3 prostate cancer and MDA-MB-231 breast cancer cells with these membrane-permeable Zn-chelators with different Zn affinities results in varying degrees of XIAP depletion. Following decreased level of XIAP expression, we also show apoptosis-related caspase activation and cellular morphological changes upon treatment with strong Zn-chelators N4Py and BnTPEN. Addition of Zn has a full protective effect on the cells treated with these chelators, while iron (Fe) addition has only partial protection that, however, can be further increased to a comparable level of protection as Zn by inhibition of ROS generation, indicating that cell death effects mediated by Fe- but not Zn-complexes involve redox cycling. These findings suggest that strong Zn-chelating agents may be useful in the treatment of apoptosis-resistant human cancers.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Próstata/metabolismo , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Zinc/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Quelantes/farmacología , Femenino , Humanos , Masculino , Estabilidad Proteica/efectos de los fármacos , Piridinas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA