Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Pharm ; 18(4): 1690-1698, 2021 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-33734721

RESUMEN

The urokinase plasminogen activator (uPA) and its cofactors are important regulators of tumor initiation and progression (including metastasis), and its overexpression is associated with unfavorable situations in cancer patients. We have previously used positron emission tomography (PET) imaging with a radiolabeled monoclonal antibody against the uPA (named ATN-291) to detect the uPA signaling activity in various cancer types; however, good tumor contrast can only be observed 24 h postinjection. To shorten the antibody circulation time and decrease interactions of ATN-291 with the mononuclear phagocyte system (MPS), our goal in this study is to develop an engineered antibody fragment (F(ab')2) from the parent antibody. By pepsin digestion and chromatography purification, ATN-291 F(ab')2 was obtained and characterized. Subsequently, it was conjugated with NOTA-Bn-NCS or fluorescein isothiocyanate (FITC) for PET imaging and fluorescence-mediated cellular analysis (i.e., flow cytometry or fluorescence microscopy). We confirmed that ATN-291 F(ab')2 still maintained a good targeting efficacy for the uPA in MDA-MB-231 cells (uPA+) and it had a faster blood clearance speed compared with ATN-291, while its interaction with MPS has been significantly decreased. In rodent tumor xenografts, radiolabeled ATN-291 F(ab')2 had a selective and persistent uptake in MDA-MB-231 tumors, with an early tumor-to-blood ratio of 1.3 ± 0.8 (n = 4) at 2 h postinjection from PET imaging. During our observation, radiolabeled ATN-291 F(ab')2 was excreted from both renal and hepatobiliary pathways. Radiolabeled ATN-291 F(ab')2 was also used for detecting uPA fluctuation during the tumor treatment in test animals. We concluded that radiolabeled ATN-291 F(ab')2 could be used as fast as PET cancer diagnostics with versatile applicability.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Fragmentos Fab de Inmunoglobulinas/administración & dosificación , Proteínas de la Membrana/antagonistas & inhibidores , Tomografía de Emisión de Positrones/métodos , Neoplasias de la Mama Triple Negativas/diagnóstico , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/genética , Femenino , Fluoresceína-5-Isotiocianato/química , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/genética , Proteínas de la Membrana/metabolismo , Ratones , Ingeniería de Proteínas , Neoplasias de la Mama Triple Negativas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Mol Pharm ; 14(3): 770-780, 2017 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-28135101

RESUMEN

High overexpression of sigma (σ) receptors (σ1 and σ2 subtypes) in a variety of human solid tumors has prompted the development of σ receptor-targeting radioligands, as imaging agents for tumor detection. A majority of these radioligands to date target the σ2 receptor, a potential marker of tumor proliferative status. The identification of approximately equal proportions of both σ receptor subtypes in prostate tumors suggests that a high affinity, dual σ receptor-targeting radioligand could potentially provide enhanced tumor targeting efficacy in prostate cancer. To accomplish this goal, we designed a series of ligands which bind to both σ receptor subtypes with high affinity. Ligand 3a in this series, displaying optimal dual σ receptor subtype affinity (σ1, 6.3 nM; σ2, 10.2 nM) was radiolabeled with fluorine-18 (18F) to give [18F]3a and evaluated as a σ receptor-targeting radioligand in the mouse PC-3 prostate tumor model. Cellular assays with PC-3 cells demonstrated that a major proportion of [18F]3a was localized to cell surface σ receptors, while ∼10% of [18F]3a was internalized within cells after incubation for 3.5 h. Serial PET imaging in mice bearing PC-3 tumors revealed that uptake of [18F]3a was 1.6 ± 0.8, 4.4 ± 0.3, and 3.6 ± 0.6% ID/g (% injection dose per gram) in σ receptor-positive prostate tumors at 15 min, 1.5 h, and 3.5 h postinjection, respectively (n = 3) resulting in clear tumor visualization. Blocking studies conducted with haloperidol (a nonselective inhibitor for both σ receptor subtypes) confirmed that the uptake of [18F]3a was σ receptor-mediated. Histology analysis confirmed similar expression of σ1 and σ2 in PC-3 tumors which was significantly greater than its expression in normal organs/tissues such as liver, kidney, and muscle. Metabolite studies revealed that >50% of radioactivity in PC-3 tumors at 30 min postinjection represented intact [18F]3a. Prominent σ receptor-specific uptake of [18F]3a in prostate tumors and its subsequent clear visualization with PET imaging indicate potential utility for the diagnosis of prostate carcinoma.


Asunto(s)
Benzamidas/farmacología , Radioisótopos de Flúor/química , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/tratamiento farmacológico , Radiofármacos/farmacología , Receptores sigma/metabolismo , Animales , Benzamidas/química , Benzamidas/metabolismo , Línea Celular Tumoral , Ligandos , Masculino , Ratones , Ratones Desnudos , Tomografía de Emisión de Positrones/métodos , Neoplasias de la Próstata/metabolismo , Radioquímica/métodos , Radiofármacos/química , Radiofármacos/metabolismo , Ratas , Distribución Tisular
3.
Bioorg Med Chem ; 23(7): 1386-94, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25766633

RESUMEN

The MAP kinase (Ras/MEK/ERK) and PI3K/Akt/mTOR oncogenic signaling pathways are central regulators of KRAS-mediated transformation. Molecular reciprocity between the Ras/MEK/ERK and PI3K/Akt/mTOR pathways provides cancer cells with the ability to evade treatment when targeting only one pathway with monotherapy. Multi-kinase targeting was explored through the development of a single bivalent chemical entity by covalent linking of high-affinity MEK and PI3K inhibitors. A prototype dual-acting agent (compound 8) designed using the PI3K inhibitor ZSTK474 and the Raf/MEK inhibitor RO5126766 as scaffolds displayed high in vitro inhibition of both PI3K (IC50=172nM) and MEK1 (IC50=473nM). Additionally, compound 8 demonstrated significant modulation of MEK and PI3K signaling pathway activity in human A549 human lung adenocarcinoma cells and pancreatic cancer cells (PANC-1) and also decreased cellular viability in these two cell lines.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/química , MAP Quinasa Quinasa 1/antagonistas & inhibidores , Inhibidores de las Quinasa Fosfoinosítidos-3 , Regulación Alostérica/efectos de los fármacos , Animales , Línea Celular , Cumarinas/administración & dosificación , Cumarinas/química , Cristalografía por Rayos X , Humanos , MAP Quinasa Quinasa 1/metabolismo , Ratones , Fosfatidilinositol 3-Quinasa/metabolismo , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Triazinas/administración & dosificación , Triazinas/química
4.
Eur J Med Chem ; 229: 113996, 2022 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-34802837

RESUMEN

Established roles for PI3K and MAPK signaling pathways in tumorigenesis has prompted extensive research towards the discovery of small-molecule inhibitors as cancer therapeutics. However, significant compensatory regulation exists between these two signaling cascades, leading to redundancy among survival pathways. Consequently, initial clinical trials aimed at either PI3K or MEK inhibition alone have proven ineffective and highlight the need for development of targeted and innovative therapeutic combination strategies. We designed a series of PI3K inhibitor derivatives wherein a single morpholine group of the PI3K inhibitor ZSTK474 was substituted with a variety of 2-aminoethyl functional groups. Analogs with pendant hydroxyl or methoxy groups maintained low nanomolar inhibition towards PI3Kα, PI3Kγ, and PI3Kδ isoforms in contrast to those with pendant amino groups which were significantly less inhibitory. Synthesis of prototype PI3K/MEK bifunctional inhibitors (6r, 6s) was guided by the structure-activity data, where a MEK-targeting inhibitor was tethered directly via a short PEG linker to the triazine core of the PI3K inhibitor analogs. These compounds (6r, 6s) displayed nanomolar inhibition towards PI3Kα, δ, and MEK (IC50 ∼105-350 nM), and low micromolar inhibition for PI3Kß and PI3Kγ (IC50 ∼1.5-3.9 µM) in enzymatic inhibition assays. Cell viability assays demonstrated superior anti-proliferative activity for 6s over 6r in three tumor-derived cell lines (A375, D54, SET-2), which correlated with inhibition of downstream AKT and ERK1/2 phosphorylation. Compounds 6r and 6s also demonstrated in vivo tolerability with therapeutic efficacy through reduction of kinase activation and amelioration of disease phenotypes in the JAK2V617F mutant myelofibrosis mouse cancer model. Taken together, these results support further structure optimization of 6r and 6s as promising leads for combination therapy in human cancer as a new class of PI3K/MEK bifunctional inhibitors.


Asunto(s)
Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Morfolinas/química , Fosfatidilinositol 3-Quinasas/química , Inhibidores de las Quinasa Fosfoinosítidos-3/química , Triazinas/química , Animales , Sitios de Unión , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Simulación del Acoplamiento Molecular , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3/uso terapéutico , Mielofibrosis Primaria/tratamiento farmacológico , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/metabolismo , Relación Estructura-Actividad , Triazinas/metabolismo , Triazinas/uso terapéutico
5.
Nat Commun ; 13(1): 4730, 2022 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-35977945

RESUMEN

Activation of compensatory signaling nodes in cancer often requires combination therapies that are frequently plagued by dose-limiting toxicities. Intestinal lymphatic drug absorption is seldom explored, although reduced toxicity and sustained drug levels would be anticipated to improve systemic bioavailability. A potent orally bioavailable multi-functional kinase inhibitor (LP-182) is described with intrinsic lymphatic partitioning for the combined targeting of phosphoinositide 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) signaling pathways without observable toxicity. We demonstrate selectivity and therapeutic efficacy through reduction of downstream kinase activation, amelioration of disease phenotypes, and improved survival in animal models of myelofibrosis. Our further characterization of synthetic and physiochemical properties for small molecule lymphatic uptake will support continued advancements in lymphatropic therapy for altering disease trajectories of a myriad of human disease indications.


Asunto(s)
Antineoplásicos , Mielofibrosis Primaria , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Humanos , Sistema de Señalización de MAP Quinasas , Fosfatidilinositol 3-Quinasas/metabolismo , Mielofibrosis Primaria/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico
6.
ACS Appl Mater Interfaces ; 11(24): 21343-21352, 2019 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-31140277

RESUMEN

Water-soluble gadofullerene nanomaterials have been extensively investigated as magnetic resonance imaging (MRI) contrast agents, radical scavengers, sensitizers for photodynamic therapy, and inherent antineoplastic agents. Most recently, an alanine-modified gadofullerene nanoparticle (Gd@C82-Ala) with excellent anticancer activity has been reported; however, the absolute tumor uptake of Gd@C82-Ala is still far from being satisfactory, and its dynamic pharmacokinetics and long-term metabolic behaviors remain to be elucidated. Herein, Gd@C82-Ala was chemically modified with eight-arm polyethylene glycol amine to improve its biocompatibility and provide the active sites for the attachment of a tumor-homing ligand (cRGD) and positron emission tomography (PET) isotopes (i.e., 64Cu or 89Zr). The physical and chemical properties (e.g., size, surface functionalization condition, radiochemical stability, etc.) of functionalized Gd@C82-Ala were properly characterized. Also, its glioblastoma cell targeting capacity was evaluated in vitro by flow cytometry, confocal fluorescence microscopy, and dynamic cellular interaction assays. Because of the presence of gadolinium ions, the gadofullerene conjugates can act simultaneously as T1* MRI contrast agents and PET probes. Thus, the pharmacokinetic behavior of functionalized Gd@C82-Ala was investigated by PET/MRI, which combines the merits of high resolution and excellent sensitivity. The functionalized Gd@C82-Ala-PEG-cRGD-NOTA-64Cu (NOTA stands for 1,4,7-triazacyclononane-triacetic acid) demonstrated much higher accumulation in U87-MG tumor than its counterpart without cRGD attachment from in vivo PET observation, consistent with observation at the cellular level. In addition, Gd@C82-Ala-PEG-Df-89Zr (Df stands for desferrioxamine) was employed to investigate the metabolic behavior of gadofullerene conjugates in vivo for up to 30 days. It was estimated that nearly 70% of Gd@C82-Ala-PEG-Df-89Zr was excreted from the test subjects primarily through renal pathways within 24 h. With proper surface engineering, functionalized Gd@C82-Ala nanoparticles can show an improved accumulation in glioblastoma. Pharmacokinetic studies also confirmed the safety of this nanoplatform, which can be used as an image-guidable therapeutic agent for glioblastoma.


Asunto(s)
Glioblastoma/diagnóstico por imagen , Imagen por Resonancia Magnética/métodos , Nanopartículas/química , Tomografía de Emisión de Positrones/métodos , Animales , Citometría de Flujo , Fulerenos/química , Gadolinio/química , Humanos , Células MCF-7 , Ratones , Microscopía Confocal
7.
J Ocul Pharmacol Ther ; 34(6): 477-485, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29708810

RESUMEN

PURPOSE: ST-162 and ST-168 are small-molecule bifunctional inhibitors of MEK and PI3K signaling pathways that are being developed as novel antitumor agents. Previous small-molecule and biologic MEK inhibitors demonstrated ocular toxicity events that were dose limiting in clinical studies. We evaluated in vitro and in vivo ocular toxicity profiles of ST-162 and ST-168. METHODS: Photoreceptor cell line 661W and adult retinal pigment epithelium cell line ARPE-19 were treated with increasing concentrations of bifunctional inhibitors. Western blots, cell viability, and caspase activity assays were performed to evaluate MEK and PI3K inhibition and dose-dependent in vitro toxicity, and compared with monotherapy. In vivo toxicity profile was assessed by intravitreal injection of ST-162 and ST-168 in Dutch-Belted rabbits, followed by ocular examination and histological analysis of enucleated eyes. RESULTS: Retinal cell lines treated with ST-162 or ST-168 exhibited dose-dependent inhibition of MEK and PI3K signaling. Compared with inhibition by monotherapies and their combinations, bifunctional inhibitors demonstrated reduced cell death and caspase activity. In vivo, both bifunctional inhibitors exhibited a more favorable toxicity profile when compared with MEK inhibitor PD0325901. CONCLUSIONS: Novel MEK and PI3K bifunctional inhibitors ST-162 and ST-168 demonstrate favorable in vitro and in vivo ocular toxicity profiles, supporting their further development as potential therapeutic agents targeting multiple aggressive tumors.


Asunto(s)
Ojo/efectos de los fármacos , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/efectos adversos , Agudeza Visual/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Humanos , Inyecciones Intravítreas , Quinasas Quinasa Quinasa PAM/metabolismo , Masculino , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacología , Conejos
8.
ACS Nano ; 11(4): 4315-4327, 2017 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-28345871

RESUMEN

Nanoscale metal-organic frameworks (nMOF) materials represent an attractive tool for various biomedical applications. Due to the chemical versatility, enormous porosity, and tunable degradability of nMOFs, they have been adopted as carriers for delivery of imaging and/or therapeutic cargos. However, the relatively low stability of most nMOFs has limited practical in vivo applications. Here we report the production and characterization of an intrinsically radioactive UiO-66 nMOF (89Zr-UiO-66) with incorporation of positron-emitting isotope zirconium-89 (89Zr). 89Zr-UiO-66 was further functionalized with pyrene-derived polyethylene glycol (Py-PGA-PEG) and conjugated with a peptide ligand (F3) to nucleolin for targeting of triple-negative breast tumors. Doxorubicin (DOX) was loaded onto UiO-66 with a relatively high loading capacity (1 mg DOX/mg UiO-66) and served as both a therapeutic cargo and a fluorescence visualizer in this study. Functionalized 89Zr-UiO-66 demonstrated strong radiochemical and material stability in different biological media. Based on the findings from cellular targeting and in vivo positron emission tomography (PET) imaging, we can conclude that 89Zr-UiO-66/Py-PGA-PEG-F3 can serve as an image-guidable, tumor-selective cargo delivery nanoplatform. In addition, toxicity evaluation confirmed that properly PEGylated UiO-66 did not impose acute or chronic toxicity to the test subjects. With selective targeting of nucleolin on both tumor vasculature and tumor cells, this intrinsically radioactive nMOF can find broad application in cancer theranostics.


Asunto(s)
Portadores de Fármacos/química , Estructuras Metalorgánicas/química , Nanopartículas/química , Neoplasias/diagnóstico por imagen , Neoplasias/terapia , Radiofármacos/química , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Línea Celular , Línea Celular Tumoral , Medios de Contraste/química , Doxorrubicina/administración & dosificación , Doxorrubicina/química , Liberación de Fármacos , Femenino , Humanos , Estructuras Metalorgánicas/toxicidad , Ratones Endogámicos BALB C , Terapia Molecular Dirigida , Péptidos/química , Polietilenglicoles/química , Tomografía de Emisión de Positrones , Radioisótopos/química , Distribución Tisular , Circonio/química
9.
ACS Med Chem Lett ; 8(8): 808-813, 2017 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-28835793

RESUMEN

The structure-based design of a new single entity, MEK/PI3K bifunctional inhibitor (7, ST-168), which displays improved MEK1 and PI3K isoform inhibition, is described. ST-168 demonstrated a 2.2-fold improvement in MEK1 inhibition and a 2.8-, 2.7-, 23-, and 2.5-fold improved inhibition toward the PI3Kα, PI3Kß, PI3Kδ, and PI3Kγ isoforms, respectively, as compared to a previous lead compound (4; ST-162) in in vitro enzymatic inhibition assays. ST-168 demonstrated superior tumoricidal efficacy over ST-162 in an A375 melanoma spheroid tumor model. ST-168 was comparatively more effective than ST-162 in promoting tumor control when administrated orally in a tumor therapy study conducted in an A375 melanoma mouse model confirming its bioavailability and efficacy toward combined in vivo MEK1/PI3K inhibition.

10.
Oncotarget ; 7(45): 73912-73924, 2016 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-27729618

RESUMEN

Mounting evidence suggests that the urokinase plasminogen activator (uPA) and its receptor (uPAR) play a central role in tumor progression. The goal of this study was to develop an 89Zr-labeled, antibody-based positron emission tomography (PET) tracer for quantitative imaging of the uPA/uPAR system. An anti-uPA monoclonal antibody (ATN-291) was conjugated with a deferoxamine (Df) derivative and subsequently labeled with 89Zr. Flow cytometry, microscopy studies, and competitive binding assays were conducted to validate the binding specificity of Df-ATN-291 against uPA. PET imaging with 89Zr-Df-ATN-291 was carried out in different tumors with distinct expression levels of uPA. Biodistribution, histology examination, and Western blotting were performed to correlate tumor uptake with uPA or uPAR expression. ATN-291 retained uPA binding affinity and specificity after Df conjugation. 89Zr-labeling of ATN-291 was achieved in good radiochemical yield and high specific activity. Serial PET imaging demonstrated that, in most tumors studied (except uPA- LNCaP), the uptake of 89Zr-Df-ATN-291 was higher compared to major organs at 120 h post-injection, providing excellent tumor contrast. The tumor-to-muscle ratio of 89Zr-Df-ATN-291 in U87MG was as high as 45.2 ± 9.0 at 120 h p.i. In vivo uPA specificity of 89Zr-Df-ATN-291 was confirmed by successful pharmacological blocking of tumor uptake with ATN-291 in U87MG tumors. Although the detailed mechanisms behind in vivo 89Zr-Df-ATN-291 tumor uptake remained to be further elucidated, quantitative PET imaging with 89Zr-Df-ATN-291 in tumors can facilitate oncologists to adopt more relevant cancer treatment planning.


Asunto(s)
Anticuerpos Monoclonales , Imagen Molecular , Neoplasias/diagnóstico por imagen , Neoplasias/metabolismo , Tomografía de Emisión de Positrones , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Animales , Biomarcadores , Línea Celular Tumoral , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Xenoinjertos , Humanos , Ratones , Imagen Molecular/métodos , Neoplasias/patología , Tomografía de Emisión de Positrones/métodos , Unión Proteica , Radiofármacos , Distribución Tisular
11.
J Med Chem ; 59(6): 2512-22, 2016 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-26943489

RESUMEN

The synthesis of a series of single entity, bifunctional MEK1/PI3K inhibitors achieved by covalent linking of structural analogs of the ATP-competitive PI3K inhibitor ZSTK474 and the ATP-noncompetitive MEK inhibitor PD0325901 is described. Inhibitors displayed potent in vitro inhibition of MEK1 (0.015 < IC50 (nM) < 56.7) and PI3K (54 < IC50 (nM) < 341) in enzymatic inhibition assays. Concurrent MEK1 and PI3K inhibition was demonstrated with inhibitors 9 and 14 in two tumor cell lines (A549, D54). Inhibitors produced dose-dependent decreased cell viability similar to the combined administration of equivalent doses of ZSTK474 and PD0325901. In vivo efficacy of 14 following oral administration was demonstrated in D54 glioma and A549 lung tumor bearing mice. Compound 14 showed a 95% and 67% inhibition of tumor ERK1/2 and Akt phosphorylation, respectively, at 2 h postadministration by Western blot analysis, confirming the bioavailability and efficacy of this bifunctional inhibitor strategy toward combined MEK1/PI3K inhibition.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , MAP Quinasa Quinasa 1/antagonistas & inhibidores , Inhibidores de las Quinasa Fosfoinosítidos-3 , Animales , Benzamidas/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Difenilamina/análogos & derivados , Difenilamina/farmacología , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Descubrimiento de Drogas , Femenino , Humanos , Ratones , Ratones Desnudos , Simulación del Acoplamiento Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Sci Signal ; 8(358): ra1, 2015 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-25564677

RESUMEN

Transforming growth factor-ß (TGF-ß) signaling regulates cell proliferation and differentiation, which contributes to development and disease. Upon binding TGF-ß, the type I receptor (TGFBRI) binds TGFBRII, leading to the activation of the transcription factors SMAD2 and SMAD3. Using an RNA interference screen of the human kinome and a live-cell reporter for TGFBR activity, we identified the kinase BUB1 (budding uninhibited by benzimidazoles-1) as a key mediator of TGF-ß signaling. BUB1 interacted with TGFBRI in the presence of TGF-ß and promoted the heterodimerization of TGFBRI and TGFBRII. Additionally, BUB1 interacted with TGFBRII, suggesting the formation of a ternary complex. Knocking down BUB1 prevented the recruitment of SMAD3 to the receptor complex, the phosphorylation of SMAD2 and SMAD3 and their interaction with SMAD4, SMAD-dependent transcription, and TGF-ß-mediated changes in cellular phenotype including epithelial-mesenchymal transition (EMT), migration, and invasion. Knockdown of BUB1 also impaired noncanonical TGF-ß signaling mediated by the kinases AKT and p38 MAPK (mitogen-activated protein kinase). The ability of BUB1 to promote TGF-ß signaling depended on the kinase activity of BUB1. A small-molecule inhibitor of the kinase activity of BUB1 (2OH-BNPP1) and a kinase-deficient mutant of BUB1 suppressed TGF-ß signaling and formation of the ternary complex in various normal and cancer cell lines. 2OH-BNPP1 administration to mice bearing lung carcinoma xenografts reduced the amount of phosphorylated SMAD2 in tumor tissue. These findings indicated that BUB1 functions as a kinase in the TGF-ß pathway in a role beyond its established function in cell cycle regulation and chromosome cohesion.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/fisiología , Proteínas Smad Reguladas por Receptores/metabolismo , Factor de Crecimiento Transformador beta/fisiología , Animales , Western Blotting , Línea Celular Tumoral , Dimerización , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Ensayos Analíticos de Alto Rendimiento , Humanos , Inmunohistoquímica , Inmunoprecipitación , Ratones , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptores de Factores de Crecimiento Transformadores beta/química , Transducción de Señal/genética
13.
PLoS One ; 6(7): e22418, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21811604

RESUMEN

A radioiodinated derivative of the tumor-homing F3 peptide, (N-(2-{3-[(125)I]Iodobenzoyl}aminoethyl)maleimide-F3Cys peptide, [(125)I]IBMF3 was developed for investigation as a SPECT tumor imaging radioligand. For this purpose, we custom synthesized a modified F3 peptide analog (F3Cys) incorporating a C-terminal cysteine residue for site-specific attachment of a radioiodinated maleimide conjugating group. Initial proof-of-concept Fluorescence studies conducted with AlexaFluor 532 C(5) maleimide-labeled F3Cys showed distinct membrane and nuclear localization of F3Cys in MDA-MB-435 cells. Additionally, F3Cys conjugated with NIR fluorochrome AlexaFluor 647 C(2) maleimide demonstrated high tumor specific uptake in melanoma cancer MDA-MB-435 and lung cancer A549 xenografts in nude mice whereas a similarly labeled control peptide did not show any tumor uptake. These results were also confirmed by ex vivo tissue analysis. No-carrier-added [(125)I]IBMF3 was synthesized by a radioiododestannylation approach in 73% overall radiochemical yield. In vitro cell uptake studies conducted with [(125)I]IBMF3 displayed a 5-fold increase in its cell uptake at 4 h when compared to controls. SPECT imaging studies with [(125)I]IBMF3 in tumor bearing nude mice showed clear visualization of MDA-MB-435 xenografts on systemic administration. These studies demonstrate a potential utility of F3 peptide-based radioligands for tumor imaging with PET or SPECT techniques.


Asunto(s)
Yodobencenos/síntesis química , Yodobencenos/metabolismo , Neoplasias/diagnóstico por imagen , Péptidos/síntesis química , Péptidos/metabolismo , Radiofármacos , Tomografía Computarizada de Emisión de Fotón Único , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Colorantes Fluorescentes/metabolismo , Humanos , Radioisótopos de Yodo , Yodobencenos/química , Ligandos , Ratones , Datos de Secuencia Molecular , Péptidos/química , Distribución Tisular
14.
Prog Mol Biol Transl Sci ; 95: 237-98, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21075334

RESUMEN

Today molecular imaging technologies play a central role in clinical oncology. The use of imaging techniques in early cancer detection, treatment response, and new therapy development is steadily growing and has already significantly impacted on clinical management of cancer. In this chapter, we overview three different molecular imaging technologies used for the understanding of disease biomarkers, drug development, or monitoring therapeutic outcome. They are (1) optical imaging (bioluminescence and fluorescence imaging), (2) magnetic resonance imaging (MRI), and (3) nuclear imaging (e.g., single-photon emission computed tomography (SPECT) and positron emission tomography (PET)). We review the use of molecular reporters of biological processes (e.g., apoptosis and protein kinase activity) for high-throughput drug screening and new cancer therapies, diffusion MRI as a biomarker for early treatment response and PET and SPECT radioligands in oncology.


Asunto(s)
Imagen Molecular/métodos , Animales , Humanos , Imagen por Resonancia Magnética , Dispositivos Ópticos , Cintigrafía
15.
Clin Cancer Res ; 16(5): 1542-52, 2010 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-20160061

RESUMEN

PURPOSE: Functional imaging biomarkers of cancer treatment response offer the potential for early determination of outcome through the assessment of biochemical, physiologic, and microenvironmental readouts. Cell death may result in an immunologic response, thus complicating the interpretation of biomarker readouts. This study evaluated the temporal effect of treatment-associated inflammatory activity on diffusion magnetic resonance imaging and 2-[(18)F]-fluoro-2-deoxy-D-glucose-positron emission tomography imaging (FDG-PET) biomarkers to delineate the effects of the inflammatory response on imaging readouts. EXPERIMENTAL DESIGN: Rats with intracerebral 9L gliosarcomas were separated into four groups consisting of control, an immunosuppressive agent dexamethasone (Dex), 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU), and BCNU+Dex. Animals were imaged using diffusion-weighted magnetic resonance imaging and FDG-PET at 0, 3, and 7 days posttreatment. RESULTS: In the BCNU- and BCNU+Dex-treated animal groups, diffusion values increased progressively over the 7-day study period to approximately 23% over baseline. The FDG percentage change of standard uptake value decreased at day 3 (-30.9%) but increased over baseline levels at day 7 (+20.1%). FDG-PET of BCNU+Dex-treated animals were found to have percentage of standard uptake value reductions of -31.4% and -24.7% at days 3 and 7, respectively, following treatment. Activated macrophages were observed on day 7 in the BCNU treatment group with much fewer found in the BCNU+Dex group. CONCLUSIONS: Results revealed that treatment-associated inflammatory response following tumor therapy resulted in the accentuation of tumor diffusion response along with a corresponding increase in tumor FDG uptake due to the presence of glucose-consuming activated macrophages. The dynamics and magnitude of potential inflammatory response should be considered when interpreting imaging biomarker results.


Asunto(s)
Antineoplásicos/efectos adversos , Neoplasias Encefálicas/patología , Imagen de Difusión por Resonancia Magnética , Inflamación/inducido químicamente , Tomografía de Emisión de Positrones , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Carmustina/efectos adversos , Dexametasona/efectos adversos , Fluorodesoxiglucosa F18 , Gliosarcoma/tratamiento farmacológico , Gliosarcoma/patología , Procesamiento de Imagen Asistido por Computador , Inflamación/patología , Radiofármacos , Ratas
16.
J Biol Chem ; 284(23): 15729-38, 2009 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-19332539

RESUMEN

Furin is a ubiquitously expressed proprotein convertase (PC) that plays a vital role in numerous disease processes including cancer metastasis, bacterial toxin activation (e.g. anthrax and Pseudomonas), and viral propagation (e.g. avian influenza and human immunodeficiency virus). To identify small molecule inhibitors of furin and related processing enzymes, we performed high-throughput screens of chemical diversity libraries utilizing both enzyme-based and cell-based assays. The screens identified partially overlapping sets of compounds that were further characterized for affinity, mechanism, and efficacy in additional cellular processing assays. Dicoumarols were identified as a class of compounds that inhibited furin non-competitively and reversibly with Ki values in the micromolar range. These compounds inhibited furin/furin-like activity both at the cell surface (protecting against anthrax toxin) and in the secretory pathway (blocking processing of the metastasis factor membrane-type 1 matrix metalloproteinase/MT1-MMP) at concentrations close to Ki values. Compounds tested exhibited distinct patterns of inhibition of other furin-family PCs (rat PACE4, human PC5/6 and human PC7), showing that dicoumarol derivatives might be developed as either generic or selective inhibitors of the PCs. The extensive clinical use, high bioavailability and relatively low toxicity of dicoumarols suggests that the dicoumarol structure will be a good starting point for development of drug-like inhibitors of furin and other PCs that can act both intracellularly and at the cell surface.


Asunto(s)
Furina/metabolismo , Proproteína Convertasas/metabolismo , Animales , Antígenos Bacterianos/toxicidad , Toxinas Bacterianas/toxicidad , Células CHO/efectos de los fármacos , Dominio Catalítico , Cricetinae , Cricetulus , Dicumarol/farmacología , Inhibidores Enzimáticos/farmacología , Furina/antagonistas & inhibidores , Furina/genética , Humanos , Cinética , Proproteína Convertasas/antagonistas & inhibidores , Proproteína Convertasas/genética , Transfección
17.
Curr Comput Aided Drug Des ; 4(1): 46-53, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19809593

RESUMEN

Spiraling drug developmental costs and lengthy time-to-market introduction are two critical challenges facing the pharmaceutical industry. The clinical trials success rate for oncology drugs is reported to be 5% as compared to other therapeutic categories (11%) with most failures often encountered late in the clinical development process. PET and SPECT nuclear imaging technologies could play an important role in facilitating the drug development process improving the speed, efficiency and cost of drug development. This review will focus on recent studies of PET and SPECT radioligands in oncology and their application in the investigation of tumor biology. The use of clinically-validated radioligands as imaging-based biomarkers in oncology could significantly impact new cancer therapeutic development.

18.
Mol Imaging ; 7(4): 187-97, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19123989

RESUMEN

The synthesis and investigation of 5-[125I]iodoindol-3-yl-beta-d-galactopyranoside ([125I]IBDG) as a radioligand for single-photon emission computed tomography (SPECT) imaging of b-galactosidase expression are described. No-carrier-added [125I]IBDG was synthesized by a radioiododestannylation approach in > 75% overall radiochemical yield and > 99% radiochemical purity. [125I]IBDG was evaluated as a substrate using beta-galactosidase-expressing (D54L) and nonexpressing (D54) human glioma cell lines. A 24-hour incubation of this substrate with cultured cells revealed a 6.5-fold greater intracellular trapping of radioactivity in D54L cells compared with D54 cells. Systemic delivery of [125I]IBDG in nude mice bearing D54L tumors failed to show significant trapping of radioactivity within these tumors by SPECT imaging. In contrast, intratumoral injection of the substrate resulted in efficient trapping of radioactivity in D54L tumors but not D54 tumors, resulting in clear SPECT visualization of the former tumor. Based on dynamic SPECT imaging and blood metabolite analysis, we conclude that although [125I]IBDG is an efficient in vivo substrate for beta-galactosidase, its rapid renal clearance hampers its intratumoral availability on systemic administration.


Asunto(s)
Galactosa/metabolismo , Galactósidos/síntesis química , Radioisótopos de Yodo/metabolismo , Ensayo de Unión Radioligante , Tomografía Computarizada de Emisión de Fotón Único/métodos , beta-Galactosidasa/metabolismo , Animales , Línea Celular Tumoral , Galactósidos/metabolismo , Expresión Génica , Humanos , Radioisótopos de Yodo/sangre , Tasa de Depuración Metabólica , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Imagen de Cuerpo Entero
19.
Bioorg Med Chem Lett ; 14(21): 5285-8, 2004 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-15454212

RESUMEN

A series of side-chain derivatives of the arylhydantoin RU 58841 and the arylthiohydantoin RU 59063, wherein the aromatic trifluoromethyl group was replaced with iodine, was synthesized for possible development as radioiodinated androgen receptor (AR) ligands. Derivatives containing the cyanomethyl, methoxyethyl and propenyl side-chains displayed moderately high affinity (K(i)=20-59nM) towards the rat AR. Side-chains containing bulky lipophilic groups such as, benzyl and phenylpropyl, were poorly tolerated (K(i)>219nM). Superior AR binding affinities (0.71nM

Asunto(s)
Hidantoínas/síntesis química , Receptores Androgénicos/metabolismo , Animales , Unión Competitiva , Hidantoínas/farmacología , Imidazoles/síntesis química , Imidazoles/farmacología , Técnicas In Vitro , Radioisótopos de Yodo , Ligandos , Masculino , Nitrilos/síntesis química , Nitrilos/farmacología , Próstata/metabolismo , Ensayo de Unión Radioligante , Ratas , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA