Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 159(7): 1591-602, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25525877

RESUMEN

Acetyl-CoA represents a central node of carbon metabolism that plays a key role in bioenergetics, cell proliferation, and the regulation of gene expression. Highly glycolytic or hypoxic tumors must produce sufficient quantities of this metabolite to support cell growth and survival under nutrient-limiting conditions. Here, we show that the nucleocytosolic acetyl-CoA synthetase enzyme, ACSS2, supplies a key source of acetyl-CoA for tumors by capturing acetate as a carbon source. Despite exhibiting no gross deficits in growth or development, adult mice lacking ACSS2 exhibit a significant reduction in tumor burden in two different models of hepatocellular carcinoma. ACSS2 is expressed in a large proportion of human tumors, and its activity is responsible for the majority of cellular acetate uptake into both lipids and histones. These observations may qualify ACSS2 as a targetable metabolic vulnerability of a wide spectrum of tumors.


Asunto(s)
Acetato CoA Ligasa/metabolismo , Acetatos/metabolismo , Neoplasias/metabolismo , Acetato CoA Ligasa/análisis , Acetato CoA Ligasa/genética , Acetilcoenzima A/metabolismo , Animales , Humanos , Inmunohistoquímica , Neoplasias Hepáticas/metabolismo , Ratones , Neoplasias/química , Neoplasias/patología , Tomografía de Emisión de Positrones , Neoplasias de la Mama Triple Negativas/química , Neoplasias de la Mama Triple Negativas/patología
2.
Nature ; 593(7858): 282-288, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33828302

RESUMEN

Cancer cells characteristically consume glucose through Warburg metabolism1, a process that forms the basis of tumour imaging by positron emission tomography (PET). Tumour-infiltrating immune cells also rely on glucose, and impaired immune cell metabolism in the tumour microenvironment (TME) contributes to immune evasion by tumour cells2-4. However, whether the metabolism of immune cells is dysregulated in the TME by cell-intrinsic programs or by competition with cancer cells for limited nutrients remains unclear. Here we used PET tracers to measure the access to and uptake of glucose and glutamine by specific cell subsets in the TME. Notably, myeloid cells had the greatest capacity to take up intratumoral glucose, followed by T cells and cancer cells, across a range of cancer models. By contrast, cancer cells showed the highest uptake of glutamine. This distinct nutrient partitioning was programmed in a cell-intrinsic manner through mTORC1 signalling and the expression of genes related to the metabolism of glucose and glutamine. Inhibiting glutamine uptake enhanced glucose uptake across tumour-resident cell types, showing that glutamine metabolism suppresses glucose uptake without glucose being a limiting factor in the TME. Thus, cell-intrinsic programs drive the preferential acquisition of glucose and glutamine by immune and cancer cells, respectively. Cell-selective partitioning of these nutrients could be exploited to develop therapies and imaging strategies to enhance or monitor the metabolic programs and activities of specific cell populations in the TME.


Asunto(s)
Neoplasias/metabolismo , Neoplasias/patología , Nutrientes/metabolismo , Microambiente Tumoral , Animales , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Femenino , Glucosa/metabolismo , Glutamina/metabolismo , Humanos , Metabolismo de los Lípidos , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Células Mieloides/inmunología , Células Mieloides/metabolismo , Neoplasias/inmunología , Microambiente Tumoral/inmunología
3.
Radiology ; 303(3): 620-631, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35191738

RESUMEN

Background The PET tracer (4S)-4-(3-[18F]fluoropropyl)-l-glutamate (18F-FSPG) targets the system xC- cotransporter, which is overexpressed in various tumors. Purpose To assess the role of 18F-FSPG PET/CT in intracranial malignancies. Materials and Methods Twenty-six patients (mean age, 54 years ± 12; 17 men; 48 total lesions) with primary brain tumors (n = 17) or brain metastases (n = 9) were enrolled in this prospective, single-center study (ClinicalTrials.gov identifier: NCT02370563) between November 2014 and March 2016. A 30-minute dynamic brain 18F-FSPG PET/CT scan and a static whole-body (WB) 18F-FSPG PET/CT scan at 60-75 minutes were acquired. Moreover, all participants underwent MRI, and four participants underwent fluorine 18 (18F) fluorodeoxyglucose (FDG) PET imaging. PET parameters and their relative changes were obtained for all lesions. Kinetic modeling was used to estimate the 18F-FSPG tumor rate constants using the dynamic and dynamic plus WB PET data. Imaging parameters were correlated to lesion outcomes, as determined with follow-up MRI and/or pathologic examination. The Mann-Whitney U test or Student t test was used for group mean comparisons. Receiver operating characteristic curve analysis was used for performance comparison of different decision measures. Results 18F-FSPG PET/CT helped identify all 48 brain lesions. The mean tumor-to-background ratio (TBR) on the whole-brain PET images at the WB time point was 26.6 ± 24.9 (range: 2.6-150.3). When 18F-FDG PET was performed, 18F-FSPG permitted visualization of non-18F-FDG-avid lesions or allowed better lesion differentiation from surrounding tissues. In participants with primary brain tumors, the predictive accuracy of the relative changes in influx rate constant Ki and maximum standardized uptake value to discriminate between poor and good lesion outcomes were 89% and 81%, respectively. There were significant differences in the 18F-FSPG uptake curves of lesions with good versus poor outcomes in the primary brain tumor group (P < .05) but not in the brain metastases group. Conclusion PET/CT imaging with (4S)-4-(3-[18F]fluoropropyl)-l-glutamate (18F-FSPG) helped detect primary brain tumors and brain metastases with a high tumor-to-background ratio. Relative changes in 18F-FSPG uptake with multi-time-point PET appear to be helpful in predicting lesion outcomes. Clinical trial registration no. NCT02370563 © RSNA, 2022 Online supplemental material is available for this article.


Asunto(s)
Neoplasias Encefálicas , Tomografía Computarizada por Tomografía de Emisión de Positrones , Neoplasias Encefálicas/diagnóstico por imagen , Fluorodesoxiglucosa F18 , Ácido Glutámico , Humanos , Masculino , Persona de Mediana Edad , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Tomografía de Emisión de Positrones/métodos , Estudios Prospectivos , Radiofármacos
4.
Bioconjug Chem ; 28(4): 1016-1023, 2017 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-28156095

RESUMEN

Translocator protein (TSPO) is a validated target for molecular imaging of a variety of human diseases and disorders. Given its involvement in cholesterol metabolism, TSPO expression is commonly elevated in solid tumors, including glioma, colorectal cancer, and breast cancer. TSPO ligands capable of detection by optical imaging are useful molecular tracers for a variety of purposes that range from quantitative biology to drug discovery. Leveraging our prior optimization of the pyrazolopyrimidine TSPO ligand scaffold for cancer imaging, we report herein a new generation of TSPO tracers with superior binding affinity and suitability for optical imaging and screening. In total, seven candidate TSPO tracers were synthesized and vetted in this study; the most promising tracer identified (29, Kd = 0.19 nM) was the result of conjugating a high-affinity TSPO ligand to a fluorophore used routinely in biological sciences (FITC) via a functional carbon linker of optimal length. Computational modeling suggested that an n-alkyl linker of eight carbons in length allows for positioning of the bulky fluorophore distal to the ligand binding domain and toward the solvent interface, minimizing potential ligand-protein interference. Probe 29 was found to be highly suitable for in vitro imaging of live TSPO-expressing cells and could be deployed as a ligand screening and discovery tool. Competitive inhibition of probe 29 quantified by fluorescence and 3H-PK11195 quantified by traditional radiometric detection resulted in equivalent affinity data for two previously reported TSPO ligands. This study introduces the utility of TSPO ligand 29 for in vitro imaging and screening and provides a structural basis for the development of future TSPO imaging ligands bearing bulky signaling moieties.


Asunto(s)
Receptores de GABA/análisis , Animales , Línea Celular Tumoral , Humanos , Ligandos , Microscopía Confocal , Modelos Moleculares , Imagen Molecular , Imagen Óptica , Unión Proteica , Ratas , Receptores de GABA/metabolismo
5.
Bioorg Med Chem Lett ; 26(15): 3472-7, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27353534

RESUMEN

Translocator protein (TSPO) represents an attractive target for molecular imaging and therapy due to its prevalence and critical roles played in oncology and other pathologies. Based upon our previously optimized pyrazolopyrimidine scaffold, we elucidated new structure activity relationships related to N,N-disubstitutions of the terminal acetamide on pyrazolopyrimidines and further explored the impacts of these substituents on lipophilicity and plasma protein binding. Several novel chemical probes reported here exhibited significantly increased binding affinity, suitable lipophilicity and protein binding compared with contemporary TSPO ligands. We illustrate that N,N-acetamide disubstitution affords opportunities to introduce diverse chemical moieties distal to the central pyrazolopyrimidine core, without sacrificing TSPO affinity. We anticipate that further exploration of N-acetamide substitutions may yield additional TSPO ligands capable of furthering the field of precision medicine.


Asunto(s)
Acetamidas/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Receptores de GABA/metabolismo , Acetamidas/química , Relación Dosis-Respuesta a Droga , Humanos , Ligandos , Estructura Molecular , Pirazoles/síntesis química , Pirazoles/química , Pirimidinas/síntesis química , Pirimidinas/química , Relación Estructura-Actividad
6.
Bioorg Med Chem Lett ; 26(3): 1044-1047, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26750251

RESUMEN

Herein, we report the discovery of 2-amino-4-bis(aryloxybenzyl)aminobutanoic acids as novel inhibitors of ASCT2(SLC1A5)-mediated glutamine accumulation in mammalian cells. Focused library development led to two novel ASCT2 inhibitors that exhibit significantly improved potency compared with prior art in C6 (rat) and HEK293 (human) cells. The potency of leads reported here represents a 40-fold improvement over our most potent, previously reported inhibitor and represents, to our knowledge, the most potent pharmacological inhibitors of ASCT2-mediated glutamine accumulation in live cells. These and other compounds in this novel series exhibit tractable chemical properties for further development as potential therapeutic leads.


Asunto(s)
Sistema de Transporte de Aminoácidos ASC/metabolismo , Butiratos/química , Glutamina/metabolismo , Sistema de Transporte de Aminoácidos ASC/antagonistas & inhibidores , Animales , Sitios de Unión , Butiratos/metabolismo , Línea Celular , Células HEK293 , Humanos , Antígenos de Histocompatibilidad Menor , Simulación del Acoplamiento Molecular , Estructura Terciaria de Proteína , Ratas , Relación Estructura-Actividad
7.
Am J Respir Cell Mol Biol ; 53(1): 50-9, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25375039

RESUMEN

Characterization of markers that identify activated macrophages could advance understanding of inflammatory lung diseases and facilitate development of novel methodologies for monitoring disease activity. We investigated whether folate receptor ß (FRß) expression could be used to identify and quantify activated macrophages in the lungs during acute inflammation induced by Escherichia coli LPS. We found that FRß expression was markedly increased in lung macrophages at 48 hours after intratracheal LPS. In vivo molecular imaging with a fluorescent probe (cyanine 5 polyethylene glycol folate) showed that the fluorescence signal over the chest peaked at 48 hours after intratracheal LPS and was markedly attenuated after depletion of macrophages. Using flow cytometry, we identified the cells responsible for uptake of cyanine 5-conjugated folate as FRß(+) interstitial macrophages and pulmonary monocytes, which coexpressed markers associated with an M1 proinflammatory macrophage phenotype. These findings were confirmed using a second model of acute lung inflammation generated by inducible transgenic expression of an NF-κB activator in airway epithelium. Using CC chemokine receptor 2-deficient mice, we found that FRß(+) macrophage/monocyte recruitment was dependent on the monocyte chemotactic protein-1/CC chemokine receptor 2 pathway. Together, our results demonstrate that folate-based molecular imaging can be used as a noninvasive approach to detect classically activated monocytes/macrophages recruited to the lungs during acute inflammation.


Asunto(s)
Receptor 2 de Folato/metabolismo , Regulación de la Expresión Génica , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patología , Imagen Molecular , Neumonía/metabolismo , Enfermedad Aguda , Animales , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Escherichia coli/química , Colorantes Fluorescentes/farmacología , Receptor 2 de Folato/genética , Lipopolisacáridos/química , Lipopolisacáridos/toxicidad , Ratones , Ratones Noqueados , Neumonía/inducido químicamente , Neumonía/genética , Neumonía/patología , Receptores CCR2/genética , Receptores CCR2/metabolismo
8.
Bioorg Med Chem Lett ; 25(1): 113-6, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25435145

RESUMEN

Herein, we report the discovery and structure-activity relationships (SAR) of 2-substituted glutamylanilides as novel probes of the steric environment comprising the amino acid binding domain of alanine-serine-cysteine transporter subtype 2 (ASCT2). Focused library development led to three novel, highly potent ASCT2 inhibitors, with N-(2-(morpholinomethyl)phenyl)-L-glutamine exhibiting the greatest potency in a live-cell glutamine uptake assay. This level of potency represents a three-fold improvement over the most potent, previously reported inhibitor in this series, GPNA. Furthermore, this and other compounds in the series exhibit tractable chemical properties for further development as potential therapeutic leads.


Asunto(s)
Sistema de Transporte de Aminoácidos ASC/química , Sistema de Transporte de Aminoácidos ASC/metabolismo , Anilidas/química , Anilidas/metabolismo , Sistema de Transporte de Aminoácidos ASC/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Humanos , Antígenos de Histocompatibilidad Menor , Unión Proteica/fisiología , Estructura Secundaria de Proteína , Relación Estructura-Actividad
9.
Am J Physiol Gastrointest Liver Physiol ; 307(1): G16-23, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24833705

RESUMEN

Individuals with familial adenomatous polyposis (FAP) harbor a germline mutation in adenomatous polyposis coli (APC). The major clinical manifestation is development of multiple colonic tumors at a young age due to stochastic loss of the remaining APC allele. Extracolonic features, including periampullary tumors, gastric abnormalities, and congenital hypertrophy of the retinal pigment epithelium, may occur. The objective of this study was to develop a mouse model that simulates these features of FAP. We combined our Lrig1-CreERT2/+ mice with Apcfl/+ mice, eliminated one copy of Apc in leucine-rich repeats and immunoglobulin-like domains protein 1 (Lrig1)-positive (Lrig1(+)) progenitor cells with tamoxifen injection, and monitored tumor formation in the colon by colonoscopy and PET. Initial loss of one Apc allele in Lrig1(+) cells results in a predictable pattern of preneoplastic changes, culminating in multiple distal colonic tumors within 50 days of induction, as well as the extracolonic manifestations of FAP mentioned above. We show that tumor formation can be monitored by noninvasive PET imaging. This inducible stem cell-driven model recapitulates features of FAP and offers a tractable platform on which therapeutic interventions can be monitored over time by colonoscopy and noninvasive imaging.


Asunto(s)
Poliposis Adenomatosa del Colon/metabolismo , Colon/metabolismo , Genes APC , Glicoproteínas de Membrana/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Lesiones Precancerosas/metabolismo , Poliposis Adenomatosa del Colon/diagnóstico por imagen , Poliposis Adenomatosa del Colon/genética , Poliposis Adenomatosa del Colon/patología , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Colon/diagnóstico por imagen , Colon/patología , Colonoscopía , Modelos Animales de Enfermedad , Hipertrofia , Glicoproteínas de Membrana/genética , Ratones , Ratones Transgénicos , Células Madre Neoplásicas/patología , Proteínas del Tejido Nervioso/genética , Tomografía de Emisión de Positrones , Lesiones Precancerosas/diagnóstico por imagen , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , Factores de Tiempo
10.
Gastroenterology ; 145(6): 1312-22.e1-8, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23993973

RESUMEN

BACKGROUND & AIMS: Chronic inflammation contributes to the pathogenesis of gastric tumorigenesis. The aurora kinase A (AURKA) gene is frequently amplified and overexpressed in gastrointestinal cancers. We investigated the roles of AURKA in inflammation and gastric tumorigenesis. METHODS: We used quantitative real-time reverse transcription polymerase chain reaction, immunofluorescence, immunohistochemistry, luciferase reporter, immunoblot, co-immunoprecipitation, and in vitro kinase assays to analyze AGS and MKN28 gastric cancer cells. We also analyzed Tff1(-/-) mice, growth of tumor xenografts, and human tissues. RESULTS: We correlated increased expression of AURKA with increased levels of tumor necrosis factor-α and inflammation in the gastric mucosa of Tff1(-/-) mice (r = 0.62; P = .0001). MLN8237, an investigational small-molecule selective inhibitor of AURKA, reduced nuclear staining of nuclear factor-κB (NF-κB) p65 in human gastric cancer samples and mouse epithelial cells, suppressed NF-κB reporter activity, and reduced expression of NF-κB target genes that regulate inflammation and cell survival. Inhibition of AURKA also reduced growth of xenograft tumors from human gastric cancer cells in mice and reversed the development of gastric tumors in Tff1(-/-) mice. AURKA was found to regulate NF-κB activity by binding directly and phosphorylating IκBα in cells. Premalignant and malignant lesions from the gastric mucosa of patients had increased levels of AURKA protein and nuclear NF-κB, compared with healthy gastric tissue. CONCLUSIONS: In analyses of gastric cancer cell lines, human tissue samples, and mouse models, we found AURKA to be up-regulated during chronic inflammation to promote activation of NF-κB and tumorigenesis. AURKA inhibitors might be developed as therapeutic agents for gastric cancer.


Asunto(s)
Adenocarcinoma/metabolismo , Aurora Quinasa A/metabolismo , Carcinogénesis/metabolismo , Inflamación/metabolismo , Neoplasias Gástricas/metabolismo , Regulación hacia Arriba , Adenocarcinoma/patología , Animales , Aurora Quinasa A/antagonistas & inhibidores , Aurora Quinasa A/efectos de los fármacos , Azepinas/farmacología , Carcinogénesis/efectos de los fármacos , Carcinogénesis/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patología , Xenoinjertos , Humanos , Técnicas In Vitro , Ratones Noqueados , Ratones Desnudos , FN-kappa B/metabolismo , Péptidos/deficiencia , Péptidos/genética , Péptidos/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Neoplasias Gástricas/patología , Factor Trefoil-1
11.
Bioorg Med Chem Lett ; 24(18): 4466-4471, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25172419

RESUMEN

A novel synthesis of the translocator protein (TSPO) ligand 7-chloro-N,N,5-trimethyl-4-oxo-3-phenyl-3,5-dihydro-4H-pyridazino[4,5-b]indole-1-acetamide (SSR180575, 3) was achieved in four steps from commercially available starting materials. Focused structure-activity relationship development about the pyridazinoindole ring at the N3 position led to the discovery of 7-chloro-N,N,5-trimethyl-4-oxo-3(6-fluoropyridin-2-yl)-3,5-dihydro-4H-pyridazino[4,5-b]indole-1-acetamide (14), a novel ligand of comparable affinity. Radiolabeling with fluorine-18 ((18)F) yielded 7-chloro-N,N,5-trimethyl-4-oxo-3(6-[(18)F]fluoropyridin-2-yl)-3,5-dihydro-4H-pyridazino[4,5-b]indole-1-acetamide ([(18)F]-14) in high radiochemical yield and specific activity. In vivo studies of [(18)F]-14 revealed this agent as a promising probe for molecular imaging of glioma.


Asunto(s)
Acetamidas/síntesis química , Descubrimiento de Drogas , Glioma/diagnóstico , Indoles/síntesis química , Imagen Molecular , Tomografía de Emisión de Positrones , Receptores de GABA/análisis , Acetamidas/química , Acetamidas/farmacología , Animales , Humanos , Indoles/química , Indoles/farmacología , Ligandos , Masculino , Estructura Molecular , Ratas , Ratas Wistar , Receptores de GABA/biosíntesis
12.
Mol Cell Proteomics ; 11(2): M111.015222, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22147731

RESUMEN

Analysis of cellular signaling networks typically involves targeted measurements of phosphorylated protein intermediates. However, phosphoproteomic analyses usually require affinity enrichment of phosphopeptides and can be complicated by artifactual changes in phosphorylation caused by uncontrolled preanalytical variables, particularly in the analysis of tissue specimens. We asked whether changes in protein expression, which are more stable and easily analyzed, could reflect network stimulation and inhibition. We employed this approach to analyze stimulation and inhibition of the epidermal growth factor receptor (EGFR) by EGF and selective EGFR inhibitors. Shotgun analysis of proteomes from proliferating A431 cells, EGF-stimulated cells, and cells co-treated with the EGFR inhibitors cetuximab or gefitinib identified groups of differentially expressed proteins. Comparisons of these protein groups identified 13 proteins whose EGF-induced expression changes were reversed by both EGFR inhibitors. Targeted multiple reaction monitoring analysis verified differential expression of 12 of these proteins, which comprise a candidate EGFR inhibition signature. We then tested these 12 proteins by multiple reaction monitoring analysis in three other models: 1) a comparison of DiFi (EGFR inhibitor-sensitive) and HCT116 (EGFR-insensitive) cell lines, 2) in formalin-fixed, paraffin-embedded mouse xenograft DiFi and HCT116 tumors, and 3) in tissue biopsies from a patient with the gastric hyperproliferative disorder Ménétrier's disease who was treated with cetuximab. Of the proteins in the candidate signature, a core group, including c-Jun, Jagged-1, and Claudin 4, were decreased by EGFR inhibitors in all three models. Although the goal of these studies was not to validate a clinically useful EGFR inhibition signature, the results confirm the hypothesis that clinically used EGFR inhibitors generate characteristic protein expression changes. This work further outlines a prototypical approach to derive and test protein expression signatures for drug action on signaling networks.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Colorrectales/metabolismo , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Gastritis Hipertrófica/metabolismo , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias/metabolismo , Proteómica , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cetuximab , Cromatografía Liquida , Neoplasias Colorrectales/tratamiento farmacológico , Factor de Crecimiento Epidérmico/farmacología , Gastritis Hipertrófica/tratamiento farmacológico , Gefitinib , Humanos , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Fosforilación/efectos de los fármacos , Estudios Prospectivos , Quinazolinas/farmacología , Transducción de Señal/efectos de los fármacos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Trasplante Heterólogo
13.
Tetrahedron Lett ; 55(1)2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24396154

RESUMEN

A novel and highly efficient synthetic method leveraging microwave-assisted organic synthesis (MAOS) to yield di-7-azaindolylmethanes (DAIMs) is reported. Under MAOS conditions, reaction of 7-azaindole with aldehydes resulted predominantly in DAIMs, as opposed to the expected 7-azaindole addition products that form at ambient temperature. Based upon studies of different indoles and azaindoles with various aromatic and aliphatic aldehydes, we herein propose a mechanism where rapid and efficient microwave heating promotes nucleophilicity of 7-azaindoles towards the corresponding alkylidene-azaindolene intermediate to form the DAIM. This sequence provides a versatile approach to efficiently synthesize novel DAIMs that may be useful pharmaceuticals.

14.
EJNMMI Radiopharm Chem ; 9(1): 51, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38935218

RESUMEN

BACKGROUND: 4-[18F]fluorobenzyl-triphenylphosphonium ([18F]FBnTP) is a lipophilic cation PET tracer. The cellular uptake of [18F]FBnTP is correlated with oxidative phosphorylation by mitochondria, which has been associated with multiple critical diseases. To date, [18F]FBnTP has been successfully applied for imaging myocardial perfusion, assessment of severity of coronary artery stenosis, delineation of the ischemic area after transient coronary occlusion, and detection/quantification of apoptosis in various animal models. Recent preclinical and clinical studies have also expanded the possibilities of using [18F]FBnTP in oncological diagnosis and therapeutic monitoring. However, [18F]FBnTP is typically prepared through a tediously lengthy four-step, three-pot reaction and required multiple synthesizer modules; Thus, such an approach remains a challenge for this promising radiopharmaceutical to be implemented for routine clinical studies. Herein, we report an optimized one-step, one-pot automated approach to produce [18F]FBnTP through a single standard commercially-available radiosynthesizer that enables centralized production for clinical use. RESULTS: The fully automated production of [18F]FBnTP took less than 55 min with radiochemical yields ranging from 28.33 ± 13.92% (non-decay corrected), apparent molar activity of 69.23 ± 45.62 GBq/µmol, and radiochemical purities of 99.79 ± 0.41%. The formulated [18F]FBnTP solution was determined to be sterile and colorless with a pH of 4.0-6.0. Our data has indicated no observable radiolysis after 8 h from the time of final product formulation and maximum assay of 7.88 GBq. CONCLUSIONS: A simplified and cGMP-compliant radiosynthesis of [18F]FBnTP has been established on the commercially available synthesizer in high activity concentration and radiochemical purity. While the preclinical and clinical studies using [18F]FBnTP PET are currently underway, the automated approaches reported herein facilitate clinical adoption of this radiotracer and warrant centralized production of [18F]FBnTP for imaging multiple patients.

15.
Cancer J ; 30(3): 159-169, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38753750

RESUMEN

ABSTRACT: Imaging glucose metabolism with [18F]fluorodeoxyglucose positron emission tomography has transformed the diagnostic and treatment algorithms of numerous malignancies in clinical practice. The cancer phenotype, though, extends beyond dysregulation of this single pathway. Reprogramming of other pathways of metabolism, as well as altered perfusion and hypoxia, also typifies malignancy. These features provide other opportunities for imaging that have been developed and advanced into humans. In this review, we discuss imaging metabolism, perfusion, and hypoxia in cancer, focusing on the underlying biology to provide context. We conclude by highlighting the ability to image multiple facets of biology to better characterize cancer and guide targeted treatment.


Asunto(s)
Fluorodesoxiglucosa F18 , Neoplasias , Tomografía de Emisión de Positrones , Humanos , Fluorodesoxiglucosa F18/metabolismo , Neoplasias/diagnóstico por imagen , Neoplasias/metabolismo , Neoplasias/diagnóstico , Tomografía de Emisión de Positrones/métodos , Radiofármacos/metabolismo , Hipoxia/metabolismo , Hipoxia/diagnóstico por imagen
16.
Int J Cancer ; 133(12): 2769-80, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23733406

RESUMEN

The aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor that belongs to the basic-helix-loop-helix (bHLH)-Per-ARNT-Sim (PAS) superfamily of transcription factors, mediates toxic response induced by environmental chemicals such as polycyclic aromatic hydrocarbons (PAH). AhR is expressed at high levels in several human breast carcinoma cell lines in direct correlation with the degree of their malignancy. Recent studies suggest a possible role for AhR in cancer independent of PAH. Therefore, we established stable AhR knockdown cells of the human breast cancer cell line MDA-MB-231 and analyzed their tumorigenic properties in in vitro and in vivo model systems. In addition we analyzed their response to radiation and chemotherapeutic treatment. AhR knockdown attenuated these cells tumorigenic properties in vitro including proliferation, anchorage independent growth, migration and apoptosis and reduced orthotopic xenograft tumor growth and lung metastasis in vivo. Notably, we observed that AhR knockdown enhanced radiation-induced apoptosis as well as significantly decreased cell clonogenic survival. Furthermore, AhR knockdown in MDA-MB-231 cells sensitized them to paclitaxel treatment, evident by a decrease in the required cytotoxic dose. Subsequent analysis revealed AhR knockdown significantly reduced phosphorylation of AKT, which impacts cell proliferation and survival. Apoptosis-focused gene expression analyses revealed an altered expression of genes regulating apoptosis in MDA-MB-231 cells. Collectively, our data identify AhR as a potential novel therapeutic target in the treatment of metastatic breast cancer.


Asunto(s)
Neoplasias de la Mama/patología , Proliferación Celular , Receptores de Hidrocarburo de Aril/fisiología , Animales , Apoptosis , Ciclo Celular , Línea Celular Tumoral , Femenino , Humanos , Neoplasias Pulmonares/secundario , Ratones , Interferencia de ARN , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Receptores de Hidrocarburo de Aril/genética , Neoplasias de la Mama Triple Negativas/patología
17.
Chembiochem ; 14(12): 1494-503, 2013 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-23881799

RESUMEN

Apoptosis is irreversible programmed cell death, characterized by a cellular cascade activation of caspase 3, which subsequently degrades proteins and other components of cells with a motif sequence. Here we report a novel reporter system to detect apoptosis, growth arrest, and cell death based on controlled and self-amplified protein degradation. The key element of the reporter system is an apoptotic sensor chimerical protein which consists of three components: procaspase 3, ubiquitin (Ub), and a strong consensus sequence of N-degron. Between each of these units is a DEVD (Asp-Glu-Val-Asp) sequence, which acts as the cleavage target of caspase 3. This non-conventional signal loss approach is much more sensitive than other native methods that are based on signal gain. The superior sensitivity is demonstrated by its effective application in 386-well high-throughput screening (HTS) with low drug concentrations and a short incubation time. The HTS selection process using this reporter system is very simple and economic. The simplicity eliminates potential errors introduced by multiple steps; there is no need for any substrate. Furthermore, the cells in the assay need not be disrupted, and the morphology of the cells can provide additional information on mechanisms. After HTS, the intact cells can also be used for other analytic analysis. This system thus has a potentially important role in the discovery and development of new anticancer drugs. It also appears to be very versatile, can be used both in vitro and in vivo with different linked reporter genes, and can be used for a variety of imaging applications.


Asunto(s)
Antineoplásicos/farmacología , Descubrimiento de Drogas , Ensayos de Selección de Medicamentos Antitumorales , Genes Reporteros/efectos de los fármacos , Apoptosis , Bioensayo , Línea Celular , Forma de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Microscopía Confocal , Proteínas Recombinantes/genética , Reproducibilidad de los Resultados
18.
EJNMMI Radiopharm Chem ; 8(1): 15, 2023 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-37486582

RESUMEN

BACKGROUND: (S)-4-(3-18F-Fluoropropyl)-L-Glutamic Acid ([18F]FSPG) is a positron emission tomography (PET) tracer that specifically targets the cystine/glutamate antiporter (xc-), which is frequently overexpressed in cancer and several neurological disorders. Pilot studies examining the dosimetry and biodistribution of [18F]FSPG in healthy volunteers and tumor detection in patients with non-small cell lung cancer, hepatocellular carcinoma, and brain tumors showed promising results. In particular, low background uptake in the brain, lung, liver, and bowel was observed that further leads to excellent imaging contrasts of [18F]FSPG PET. However, reliable production-scale cGMP-compliant automated procedures for [18F]FSPG production are still lacking to further increase the utility and clinical adoption of this radiotracer. Herein, we report the optimized automated approaches to produce [18F]FSPG through two commercially available radiosynthesizers capable of supporting centralized and large-scale production for clinical use. RESULTS: Starting with activity levels of 60-85 GBq, the fully-automated process to produce [18F]FSPG took less than 45 min with average radiochemical yields of 22.56 ± 0.97% and 30.82 ± 1.60% (non-decay corrected) using TRACERlab™ FXFN and FASTlab™, respectively. The radiochemical purities were > 95% and the formulated [18F]FSPG solution was determined to be sterile and colorless with the pH of 6.5-7.5. No radiolysis of the product was observed up to 8 h after final batch formulation. CONCLUSIONS: In summary, cGMP-compliant radiosyntheses and quality control of [18F]FSPG have been established on two commercially available synthesizers leveraging high activity concentration and radiochemical purity. While the clinical trials using [18F]FSPG PET are currently underway, the automated approaches reported herein will accelerate the clinical adoption of this radiotracer and warrant centralized and large-scale production of [18F]FSPG.

19.
Tomography ; 9(2): 497-508, 2023 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-36961000

RESUMEN

Early response assessment is critical for personalizing cancer therapy. Emerging therapeutic regimens with encouraging results in the wild-type (WT) KRAS colorectal cancer (CRC) setting include inhibitors of epidermal growth factor receptor (EGFR) and glutaminolysis. Towards predicting clinical outcome, this preclinical study evaluated non-invasive positron emission tomography (PET) with (4S)-4-(3-[18F]fluoropropyl)-L-glutamic acid ([18F]FSPG) in treatment-sensitive and treatment-resistant WT KRAS CRC patient-derived xenografts (PDXs). Tumor-bearing mice were imaged with [18F]FSPG PET before and one week following the initiation of treatment with either EGFR-targeted monoclonal antibody (mAb) therapy, glutaminase inhibitor therapy, or the combination. Imaging was correlated with tumor volume and histology. In PDX that responded to therapy, [18F]FSPG PET was significantly decreased from baseline at 1-week post-therapy, prior to changes in tumor volume. In contrast, [18F]FSPG PET was not decreased in non-responding PDX. These data suggest that [18F]FSPG PET may serve as an early metric of response to EGFR and glutaminase inhibition in the WT KRAS CRC setting.


Asunto(s)
Neoplasias Colorrectales , Glutaminasa , Humanos , Ratones , Animales , Glutaminasa/metabolismo , Glutamina , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Glutamatos/metabolismo , Estudios de Factibilidad , Tomografía de Emisión de Positrones/métodos , Receptores ErbB/metabolismo , Modelos Animales de Enfermedad , Neoplasias Colorrectales/diagnóstico por imagen , Neoplasias Colorrectales/tratamiento farmacológico
20.
Mol Imaging Biol ; 25(6): 991-1019, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37845582

RESUMEN

Critical advances in radionuclide therapy have led to encouraging new options for cancer treatment through the pairing of clinically useful radiation-emitting radionuclides and innovative pharmaceutical discovery. Of the various subatomic particles used in therapeutic radiopharmaceuticals, alpha (α) particles show great promise owing to their relatively large size, delivered energy, finite pathlength, and resulting ionization density. This review discusses the therapeutic benefits of α-emitting radiopharmaceuticals and their pairing with appropriate diagnostics, resulting in innovative "theranostic" platforms. Herein, the current landscape of α particle-emitting radionuclides is described with an emphasis on their use in theranostic development for cancer treatment. Commonly studied radionuclides are introduced and recent efforts towards their production for research and clinical use are described. The growing popularity of these radionuclides is explained through summarizing the biological effects of α radiation on cancer cells, which include DNA damage, activation of discrete cell death programs, and downstream immune responses. Examples of efficient α-theranostic design are described with an emphasis on strategies that lead to cellular internalization and the targeting of proteins involved in therapeutic resistance. Historical barriers to the clinical deployment of α-theranostic radiopharmaceuticals are also discussed. Recent progress towards addressing these challenges is presented along with examples of incorporating α-particle therapy in pharmaceutical platforms that can be easily converted into diagnostic counterparts.


Asunto(s)
Neoplasias , Radiofármacos , Radiofármacos/uso terapéutico , Partículas alfa/uso terapéutico , Radioisótopos/uso terapéutico , Preparaciones Farmacéuticas , Neoplasias/diagnóstico por imagen , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA