Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Mol Carcinog ; 54(7): 554-65, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24347249

RESUMEN

Hypoxic conditions in prostate cancer (PCA) are associated with poor prognosis; however, precise mechanism/s through which hypoxia promotes malignant phenotype remains unclear. Here, we analyzed the role of exosomes from hypoxic PCA cells in enhancing the invasiveness and stemness of naïve PCA cells, as well as in promoting cancer-associated fibroblast (CAF) phenotype in prostate stromal cells (PrSC). Human PCA LNCaP and PC3 cells were exposed to hypoxic (1% O2 ) or normoxic (21% O2 ) conditions, and exosomes secreted under hypoxic (Exo(Hypoxic) ) and normoxic (Exo(Normoxic) ) conditions were isolated from conditioned media. Nanoparticle tracking analysis revealed that Exo(Hypoxic) have smaller average size as compared to Exo(Normoxic) . Immunoblotting results showed a higher level of tetraspanins (CD63 and CD81), heat shock proteins (HSP90 and HSP70), and Annexin II in Exo(Hypoxic) compared to Exo(Normoxic) . Co-culturing with Exo(Hypoxic) increased the invasiveness and motility of naïve LNCaP and PC3 cells, respectively. Exo(Hypoxic) also promoted prostasphere formation by both LNCaP and PC3 cells, and enhanced α-SMA (a CAF biomarker) expression in PrSC. Compared to Exo(Normoxic) , Exo(Hypoxic) showed higher metalloproteinases activity and increased level of diverse signaling molecules (TGF-ß2, TNF1α, IL6, TSG101, Akt, ILK1, and ß-catenin). Furthermore, proteome analysis revealed a higher number of proteins in Exo(Hypoxic) (160 proteins) compared to Exo(Normoxic) (62 proteins), primarily associated with the remodeling of epithelial adherens junction pathway. Importantly, Exo(Hypoxic) targeted the expression of adherens junction proteins in naïve PC3 cells. These findings suggest that Exo(Hypoxic) are loaded with unique proteins that could enhance invasiveness, stemness, and induce microenvironment changes; thereby, promoting PCA aggressiveness.


Asunto(s)
Uniones Adherentes/patología , Exosomas/patología , Hipoxia/complicaciones , Próstata/patología , Neoplasias de la Próstata/complicaciones , Neoplasias de la Próstata/patología , Uniones Adherentes/metabolismo , Anexina A2/análisis , Anexina A2/metabolismo , Hipoxia de la Célula , Línea Celular Tumoral , Movimiento Celular , Técnicas de Cocultivo , Exosomas/metabolismo , Proteínas de Choque Térmico/análisis , Proteínas de Choque Térmico/metabolismo , Humanos , Hipoxia/metabolismo , Hipoxia/patología , Masculino , Metaloproteasas/análisis , Metaloproteasas/metabolismo , Invasividad Neoplásica/patología , Próstata/metabolismo , Neoplasias de la Próstata/metabolismo , Proteoma/análisis , Proteoma/metabolismo , Transducción de Señal , Tetraspaninas/análisis , Tetraspaninas/metabolismo
2.
Radiat Res ; 199(2): 132-147, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36583948

RESUMEN

Clonogenic assays evaluate the ability of single cells to proliferate and form colonies. This process approximates the regrowth and recurrence of tumors after treatment with radiation or chemotherapy, and thereby provides a drug discovery platform for compounds that block this process. However, because of their labor-intensive and cumbersome nature, adapting canonical clonogenic assays for high throughput screening (HTS) has been challenging. We overcame these barriers by developing an integrated system that automates cell- and liquid-handling, irradiation, dosimetry, drug administration, and incubation. Further, we developed a fluorescent live-cell based automated colony scoring methodology that identifies and counts colonies precisely based upon actual nuclei number rather than colony area, thereby eliminating errors in colony counts caused by radiation induced changes in colony morphology. We identified 13 cell lines from 7 cancer types, where radiation is a standard treatment module, that exhibit identical radiation and chemoradiation response regardless of well format and are amenable to miniaturization into small-well HTS formats. We performed pilot screens through a 1,584 compound NCI Diversity Set library using two cell lines representing different cancer indications. Radiation modulators identified in the pilot screens were validated in traditional clonogenic assays, providing proof-of-concept for the screen. The integrated methodology, hereafter "clonogenic HTS", exhibits excellent robustness (Z' values > 0.5) and shows high reproducibility (>95%). We propose that clonogenic HTS we developed can function as a drug discovery platform to identify compounds that inhibit tumor regrowth following radiation therapy, to identify new efficacious pair-wise combinations of known oncologic therapies, or to identify novel modulators ofapproved therapies.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento , Neoplasias , Humanos , Reproducibilidad de los Resultados , Línea Celular , Ensayos Analíticos de Alto Rendimiento/métodos , Descubrimiento de Drogas/métodos
3.
Genet Med ; 14(9): 811-8, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22653535

RESUMEN

PURPOSE: The aim of this study was to characterize the clinical phenotype of patients with tetrasomy of the distal 15q chromosome in the form of a neocentric marker chromosome and to evaluate whether the phenotype represents a new clinical syndrome or is a phenocopy of Shprintzen-Goldberg syndrome. METHODS: We carried out comprehensive clinical evaluation of four patients who were identified with a supernumerary marker chromosome. The marker chromosome was characterized by G-banding, fluorescence in situ hybridization, single nucleotide polymorphism oligonucleotide microarray analysis, and immunofluorescence with antibodies to centromere protein C. RESULTS: The marker chromosomes were categorized as being neocentric with all showing tetrasomy for regions distal to 15q25 and the common region of overlap being 15q26→qter. CONCLUSION: Tetrasomy of 15q26 likely results in a distinct syndrome as the patients with tetrasomy 15q26 share a strikingly more consistent phenotype than do the patients with Shprintzen-Goldberg syndrome, who show remarkable clinical variation.


Asunto(s)
Aracnodactilia/diagnóstico , Cromosomas Humanos Par 15 , Craneosinostosis/diagnóstico , Síndrome de Marfan/diagnóstico , Tetrasomía/genética , Adulto , Aracnodactilia/genética , Aracnodactilia/patología , Niño , Preescolar , Proteínas Cromosómicas no Histona/genética , Bandeo Cromosómico , Craneosinostosis/genética , Craneosinostosis/patología , Femenino , Marcadores Genéticos , Humanos , Hibridación Fluorescente in Situ , Cariotipificación , Masculino , Síndrome de Marfan/genética , Síndrome de Marfan/patología , Fenotipo , Síndrome , Tetrasomía/patología
4.
Am J Med Genet A ; 158A(7): 1574-8, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22639450

RESUMEN

We report on maternal half-sibs born to unaffected, non-consanguineous parents with classical Shprintzen-Goldberg syndrome (SGS) who had in addition intestinal malrotation and an aberrant subclavian artery. In one other SGS family germline mosaicism has been described. SGS is molecularly heterogeneous and has been linked to mutations in three genomic loci. This suggests there may be multiple other genetic factors that result in a common clinical phenotype and a number of investigators have implicated a fourth region (15q25-qter) in the etiology of SGS.


Asunto(s)
Aracnodactilia/genética , Craneosinostosis/genética , Mutación de Línea Germinal , Síndrome de Marfan/genética , Mosaicismo , Facies , Femenino , Fibrilinas , Humanos , Lactante , Masculino , Proteínas de Microfilamentos/genética , Fenotipo
5.
Radiat Res ; 194(5): 519-531, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-32936912

RESUMEN

Head and neck squamous cell carcinoma (HNSCC) is a challenging cancer with little change in five-year overall survival rate of 50-60% over the last two decades. Radiation with or without platinum-based drugs remains the standard of care despite limited benefit and high toxicity. HNSCCs often overexpress epidermal growth factor receptor (EGFR) and inhibition of EGFR signaling enhances radiation sensitivity by interfering with repair of radiation-induced DNA breaks. Poly (adenosine diphosphate-ribose) polymerase-1 (PARP1) also participates in DNA damage repair, but its inhibition provides benefit in cancers that lack DNA repair by homologous recombination (HR) such as BRCA-mutant breast cancer. HNSCCs in contrast are typically BRCA wild-type and proficient in HR repair, making it challenging to apply anti-PARP1 therapy in this model. A recently published study showed that a combination of EGFR and PARP1 inhibition induced more DNA damage and greater growth control than each single agent in HNSCC cells. This led us to hypothesize that a combination of EGFR and PARP1 inhibition would enhance the efficacy of radiation to a greater extent than each single agent, providing a rationale for paradigm-shifting combinatorial approaches to improve the standard of care in HNSCC. Here, we report a proof-of-concept study using Detroit562 HNSCC cells, which are proficient for DNA repair by both HR and non-homologous end joining (NHEJ) mechanisms. We tested the effect of adding cetuximab and/or olaparib (inhibitors of EGFR and PARP1, respectively) to radiation and compared it to that of cisplatin and radiation combination, which is the standard of care. Our results demonstrate that the combination of cetuximab and olaparib with radiation was superior to the combination of any single drug with radiation in terms of induction of unrepaired DNA damage, induction of senescence, apoptosis and clonogenic death, and tumor growth control in mouse xenografts. Combined with our recently published phase I safety data on cetuximab/olaparib/radiation triple combination, the data reported here demonstrate a potential for combining biologically-based therapies that might optimize radiosensitization in HNSCC.


Asunto(s)
Cetuximab/farmacología , Quimioterapia Adyuvante , Reparación del ADN/efectos de los fármacos , Neoplasias de Cabeza y Cuello/radioterapia , Proteínas de Neoplasias/antagonistas & inhibidores , Ftalazinas/farmacología , Piperazinas/farmacología , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Carcinoma de Células Escamosas de Cabeza y Cuello/radioterapia , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Línea Celular Tumoral , Cetuximab/administración & dosificación , Cetuximab/uso terapéutico , Cisplatino/farmacología , Cisplatino/uso terapéutico , Células Clonales , Terapia Combinada , Reparación del ADN por Unión de Extremidades , ADN de Neoplasias/efectos de los fármacos , ADN de Neoplasias/efectos de la radiación , Sinergismo Farmacológico , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Receptores ErbB/fisiología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/patología , Humanos , Ratones , Ratones Desnudos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Ftalazinas/administración & dosificación , Ftalazinas/uso terapéutico , Piperazinas/administración & dosificación , Piperazinas/uso terapéutico , Poli(ADP-Ribosa) Polimerasa-1/genética , Poli(ADP-Ribosa) Polimerasa-1/fisiología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/administración & dosificación , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Prueba de Estudio Conceptual , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Clin Cancer Res ; 26(12): 3058-3070, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32139402

RESUMEN

PURPOSE: SMAD4 loss causes genomic instability and the initiation/progression of head and neck squamous cell carcinoma (HNSCC). Here, we study whether SMAD4 loss sensitizes HNSCCs to olaparib (PARP inhibitor) in combination with radiotherapy (RT). EXPERIMENTAL DESIGN: We analyzed HNSCC The Cancer Genome Atlas data for SMAD4 expression in association with FANC/BRCA family gene expression. Human HNSCC cell lines were screened for sensitivity to olaparib. Isogenic HNSCC cell lines were generated to restore or reduce SMAD4 expression and treated with olaparib, radiation, or the combination. HNSCC pretreatment specimens from a phase I trial investigating olaparib were analyzed. RESULTS: SMAD4 levels correlated with levels of FANC/BRCA genes in HNSCC. HNSCC cell lines with SMAD4 homozygous deletion were sensitive to olaparib. In vivo, olaparib or RT monotherapy reduced tumor volumes in SMAD4-mutant but not SMAD4-positive tumors. Olaparib with RT dual therapy sustained tumor volume reduction in SMAD4-deficient (mutant or knockdown) xenografts, which exhibited increased DNA damage and cell death compared with vehicle-treated tumors. In vitro, olaparib alone or in combination with radiation caused lower clonogenic survival, more DNA damage-associated cell death, and less proliferation in SMAD4-deficient cells than in SMAD4-positive (endogenous SMAD4 or transduced SMAD4) cells. Applicable to clinic, 5 out of 6 SMAD4-negative HNSCCs and 4 out of 8 SMAD4-positive HNSCCs responded to a standard treatment plus olaparib in a phase I clinical trial, and SMAD4 protein levels inversely correlated with DNA damage. CONCLUSIONS: SMAD4 levels are causal in determining sensitivity to PARP inhibition in combination with RT in HNSCCs.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Modelos Animales de Enfermedad , Neoplasias de Cabeza y Cuello/radioterapia , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Proteína Smad4/deficiencia , Carcinoma de Células Escamosas de Cabeza y Cuello/radioterapia , Animales , Apoptosis , Proliferación Celular , Cetuximab/administración & dosificación , Femenino , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , Ratones , Ratones Desnudos , Ftalazinas/administración & dosificación , Piperazinas/administración & dosificación , Pronóstico , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Cancer Res ; 80(5): 1183-1198, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31911553

RESUMEN

Cancer stem cells (CSC) drive growth, therapy resistance, and recurrence in head and neck squamous cell carcinoma (HNSCC). Regulation of protein translation is crucial for normal stem cells and CSCs; its inhibition could disrupt stemness properties, but translation inhibitors are limited clinically due to toxicity. SVC112 is a synthetic derivative of bouvardin, a plant-derived translation elongation inhibitor. SVC112 had greater antiproliferative effects on HNSCC cells compared with the FDA-approved translation inhibitor omacetaxine mepesuccinate (HHT). SVC112 preferentially inhibited cancer cells compared with patient-matched cancer-associated fibroblasts, whereas HHT was equally toxic to both. SVC112 reduced sphere formation by cell lines and CSCs. SVC112 alone inhibited the growth of patient-derived xenografts (PDX), and SVC112 combined with radiation resulted in tumor regression in HPV-positive and HPV-negative HNSCC PDXs. Notably, CSC depletion after SVC112 correlated with tumor response. SVC112 preferentially impeded ribosomal processing of mRNAs critical for stress response and decreased CSC-related proteins including Myc and Sox2. SVC112 increased cell-cycle progression delay and slowed DNA repair following radiation, enhancing colony and sphere formation radiation effects. In summary, these data demonstrate that SVC112 suppresses CSC-related proteins, enhances the effects of radiation, and blocks growth of HNSCC PDXs by inhibiting CSCs. SIGNIFICANCE: Inhibiting protein elongation with SVC112 reduces tumor growth in head and neck squamous cell carcinoma and increases the effects of radiation by targeting the cancer stem cell pool.


Asunto(s)
Neoplasias de Cabeza y Cuello/terapia , Células Madre Neoplásicas/efectos de los fármacos , Péptidos Cíclicos/farmacología , Inhibidores de la Síntesis de la Proteína/farmacología , Carcinoma de Células Escamosas de Cabeza y Cuello/terapia , Animales , Ciclo Celular/efectos de los fármacos , Ciclo Celular/efectos de la radiación , Línea Celular Tumoral , Quimioradioterapia/métodos , Daño del ADN/efectos de la radiación , Reparación del ADN/efectos de los fármacos , Relación Dosis-Respuesta en la Radiación , Femenino , Neoplasias de Cabeza y Cuello/patología , Humanos , Ratones , Recurrencia Local de Neoplasia , Células Madre Neoplásicas/efectos de la radiación , Extensión de la Cadena Peptídica de Translación/efectos de los fármacos , Péptidos Cíclicos/química , Inhibidores de la Síntesis de la Proteína/uso terapéutico , Dosificación Radioterapéutica , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Cancer Chemother Pharmacol ; 61(2): 179-88, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17393165

RESUMEN

PURPOSE: Vandetanib (ZD6474, Zactima) is a novel, orally available inhibitor of vascular endothelial growth factor receptor-2 (VEGFR2) tyrosine kinase activity with additional activity against epidermal growth factor receptor (EGFR) tyrosine kinase. Vandetanib has demonstrated enhanced efficacy in combination with radiation therapy (RT) in human tumor models. This study aimed to evaluate the schedule-dependent interaction of clinically relevant dosing of vandetanib with RT in human head and neck cancer models that had been characterized as EGFR positive (EGFR+) or negative (EGFR-) in order to begin differentiating vandetanib and RT interactions at the level of antitumor (EGFR) or antivascular (VEGFR2) activities. METHODS: The human head and neck squamous cell carcinoma (HNSCC) cell lines UMSCC2 (EGFR+) and UMSCC10 (EGFR(-)) are sensitive and resistant to EGFR inhibitors, respectively, while having similar sensitivity to ionizing radiation. Nude mice with UMSCC2 or UMSCC10 tumor xenografts were treated with vandetanib or RT alone, or with combinations of concomitant and sequential therapy. Vandetanib was dosed at 30 mg kg(-1) day(-1) based on pharmacokinetic studies in nude mice showing that this dose results in drug exposure similar to that seen in humans at clinical doses. RT was dosed at 3 Gy twice a week for two consecutive weeks for a total dose of 12 Gy. RESULTS: Vandetanib alone caused regression in EGFR+ but not EGFR- tumors and RT therapy alone was similar in both tumor types. Combinations of vandetanib and RT showed concomitant use of vandetanib and RT was superior to RT followed by vandetanib or visa versa in EGFR- tumors. Therapeutic response of EGFR+ tumors was similar regardless of treatment sequencing. CONCLUSIONS: The combination of vandetanib and RT is active in both EGFR+ and EGFR- HNSCC tumor xenografts, however, vandetanib alone is only active in EGFR+ xenografts. EGFR+ tumor response to vandetanib and RT was independent of treatment sequencing, but concomitant treatment was superior to sequencing in EGFR- tumors. These results suggest that the anti-VEGFR2 antitumor activity of vandetanib is enhanced by RT as presumably the activity seen in EGFR- tumors is due to antiangiogenic activity, whereas the anti-EGFR antitumor activity dominates in EGFR+ tumors such that RT enhancement is not observed.


Asunto(s)
Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Neoplasias de Cabeza y Cuello/terapia , Piperidinas/administración & dosificación , Piperidinas/uso terapéutico , Quinazolinas/administración & dosificación , Quinazolinas/uso terapéutico , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Terapia Combinada , Gefitinib , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/radioterapia , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Piperidinas/efectos adversos , Quinazolinas/efectos adversos , Transducción de Señal/efectos de los fármacos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Mol Cancer Ther ; 6(6): 1683-91, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17541031

RESUMEN

The modest response of patients with head and neck squamous cell carcinoma (HNSCC) and non-small cell lung carcinoma (NSCLC) to epithelial growth factor receptor tyrosine kinase inhibitors such as gefitinib and erlotinib indicates the need for the development of biomarkers to predict response. We determined gefitinib sensitivity in a panel of HNSCC cell lines by a 5-day 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and confirmed these responses with analysis of downstream signaling by immunoblotting and cell cycle arrest. Basal gene expression profiles were then determined by microarray analysis and correlated with gefitinib response. These data were combined with previously reported NSCLC microarray results to generate a broader predictive index. Common markers of resistance between the two tumor types included genes associated with the epithelial to mesenchymal transition. We confirmed that increased protein expression of vimentin combined with the loss of E-cadherin, claudin 4, and claudin 7 by immunoblotting was associated with gefitinib resistance in both HNSCC and NSCLC cell lines. In addition, the loss of the Ca(2+)-independent cell-cell adhesion molecules EpCAM and TROP2 in resistant lines was confirmed by immunofluorescence. Tumor xenografts derived from the gefitinib-sensitive UM-SCC-2 were growth-delayed by gefitinib, whereas the gefitinib-resistant 1483 xenografts were unaffected. These data support a role for epithelial to mesenchymal transition in establishing gefitinib resistance for both HNSCC and NSCLC, and indicate that clinical trials should address whether these biomarkers will be useful for patient selection.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Células Escamosas/patología , Resistencia a Antineoplásicos , Células Epiteliales/patología , Neoplasias de Cabeza y Cuello/patología , Neoplasias Pulmonares/patología , Mesodermo/patología , Quinazolinas/farmacología , Animales , Antineoplásicos/farmacología , División Celular/efectos de los fármacos , Línea Celular Tumoral , Femenino , Gefitinib , Ratones , Ratones Desnudos , Análisis de Secuencia por Matrices de Oligonucleótidos
10.
Int J Radiat Oncol Biol Phys ; 64(1): 33-7, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16377413

RESUMEN

Radiation enhances both epithelial growth factor receptor (EGFR) and vascular endothelial growth factor (VEGF) expression, which are a part of key pathways for tumor progression. Some tumors may not respond well to EGFR inhibitors alone or may develop resistance to EGFR inhibitors. Therefore, drug therapy targeted to VEGF receptors and EGFRs, when combined with radiotherapy (RT), may improve tumor control and provide wider applicability. This article focuses on ZD6474, an inhibitor of EGFR and VEGF receptor signaling in combination with RT. We discuss preclinical and clinical studies with RT and inhibitors of VEGF or EGFR signaling first. We then address issues associated with ZD6474 pharmacokinetic dosing, and scheduling when combined with RT. We also discuss ZD6474 in the context of anti-EGFR therapy resistance. Dual inhibition of EGFR and VEGF receptor signaling pathways shows promise in enhancing RT efficacy.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Receptores ErbB/antagonistas & inhibidores , Neoplasias/radioterapia , Neovascularización Patológica/radioterapia , Piperidinas/uso terapéutico , Quinazolinas/uso terapéutico , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Inhibidores de la Angiogénesis/farmacocinética , Animales , Terapia Combinada , Receptores ErbB/metabolismo , Receptores ErbB/efectos de la radiación , Humanos , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/etiología , Neovascularización Patológica/metabolismo , Piperidinas/farmacocinética , Quinazolinas/farmacocinética , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/efectos de la radiación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Regulación hacia Arriba , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores
11.
Cancer Res ; 64(6): 2090-5, 2004 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15026348

RESUMEN

Pokeweed antiviral protein (PAP) is a plant-derived, highly potent ribosome inactivating protein that causes inhibition of protein translation and rapid cell death. We and others have delivered this protein to various cell types, including cancer cells, using hormones to specifically target cells bearing the hormone receptor. Here, we compare binding and cytotoxicity of GnRH-PAP hormonotoxins prepared either by protein conjugation (GnRH-PAP conjugate) or through recombinant DNA technology (GnRH-PAP fusion). Although GnRH-PAP conjugate protein bound specifically to and caused cell death in cells bearing the gonadotropin-releasing hormone (GnRH) receptor, we could not detect binding or cytotoxicity using two different versions of the fusion protein in receptor-positive cells. We conclude that generation of an active GnRH-PAP fusion protein may not be feasible either because both ends of the GnRH molecule are required for receptor binding, but only the NH(2) terminus is free in the fusion protein and/or that more potent analogues of GnRH (inclusion of which is not feasible in the fusion protein) are needed for efficient targeting. In contrast, the GnRH-PAP conjugate shows promise as a novel anticancer agent, capable of targeting cancer cells expressing the GnRH receptor such as prostate, breast, ovarian, endometrial, and pancreatic cells. It may also be useful as a therapeutic agent to eliminate pituitary gonadotrophs, eliminating the need for chronic GnRH analogue administration to treat hormone-sensitive diseases.


Asunto(s)
Inmunotoxinas/toxicidad , N-Glicosil Hidrolasas/metabolismo , Proteínas de Plantas/metabolismo , Receptores LHRH/metabolismo , Proteínas Recombinantes de Fusión/toxicidad , Animales , Apoptosis , Western Blotting , Células CHO , Bovinos , Membrana Celular/metabolismo , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Cricetinae , Humanos , Hipófisis/metabolismo , Unión Proteica , Biosíntesis de Proteínas/efectos de los fármacos , Proteínas Inactivadoras de Ribosomas Tipo 1
12.
Radiat Res ; 184(4): 392-403, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26414509

RESUMEN

Protein synthesis is essential for growth, proliferation and survival of cells. Translation factors are overexpressed in many cancers and in preclinical models, their experimental inhibition has been shown to inhibit cancer growth. Differential regulation of translation also occurs upon exposure to cancer-relevant stressors such as hypoxia and ionizing radiation. The failure to regulate translation has been shown to interfere with recovery after genotoxic stress. These findings suggest that modulation of translation, alone or in conjunction with genotoxins, may be therapeutic in oncology. Yet, only two drugs that directly inhibit translation are FDA-approved for oncology therapies used today. We have previously identified the protein synthesis inhibitor, bouvardin in a screen for small molecule enhancers of ionizing radiation in Drosophila melanogaster . Bouvardin was independently identified in a screen for selective inhibitors of engineered human breast cancer stem cells. Here we report the effect of bouvardin treatment in preclinical models of head and neck cancer (HNC) and glioma, two cancer types for which radiation therapy is the most common treatment. Our data show that bouvardin treatment blocked translation elongation on human ribosomes and suggest that it did so by blocking the dissociation of elongation factor 2 from the ribosome. Bouvardin and radiation enhanced the induction of clonogenic death in HNC and glioma cells, although by different mechanisms. Bouvardin treatment enhanced the radiation-induced antitumor effects in HNC tumor xenografts in mice. These data suggest that inhibition of translation elongation, particularly in combination with radiation treatment, may be a promising treatment option for cancer.


Asunto(s)
Péptidos Cíclicos/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Glioma/patología , Células HeLa , Humanos , Biosíntesis de Proteínas/efectos de los fármacos
13.
Cancer Res ; 74(23): 7024-36, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25297633

RESUMEN

Local control and overall survival in patients with advanced head and neck squamous cell cancer (HNSCC) remains dismal. Signaling through the Hedgehog (Hh) pathway is associated with epithelial-to-mesenchymal transition, and activation of the Hh effector transcription factor Gli1 is a poor prognostic factor in this disease setting. Here, we report that increased GLI1 expression in the leading edge of HNSCC tumors is further increased by irradiation, where it contributes to therapeutic inhibition. Hh pathway blockade with cyclopamine suppressed GLI1 activation and enhanced tumor sensitivity to radiotherapy. Furthermore, radiotherapy-induced GLI1 expression was mediated in part by the mTOR/S6K1 pathway. Stroma exposed to radiotherapy promoted rapid tumor repopulation, and this effect was suppressed by Hh inhibition. Our results demonstrate that Gli1 that is upregulated at the tumor-stroma intersection in HNSCC is elevated by radiotherapy, where it contributes to stromal-mediated resistance, and that Hh inhibitors offer a rational strategy to reverse this process to sensitize HNSCC to radiotherapy.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Proteínas Hedgehog/metabolismo , Tolerancia a Radiación/fisiología , Células del Estroma/metabolismo , Células del Estroma/patología , Animales , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/radioterapia , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/fisiología , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/radioterapia , Humanos , Ratones , Ratones Desnudos , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal/efectos de los fármacos , Carcinoma de Células Escamosas de Cabeza y Cuello , Células del Estroma/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Factores de Transcripción/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Alcaloides de Veratrum/farmacología , Proteína con Dedos de Zinc GLI1
14.
Mol Cancer Ther ; 12(10): 1994-2005, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23873848

RESUMEN

The dual pathway inhibitor rigosertib inhibits phosphoinositide 3-kinase (PI3K) pathway activation as well as polo-like kinase 1 (PLK1) activity across a broad spectrum of cancer cell lines. The importance of PIK3CA alterations in squamous cell carcinoma of the head and neck (HNSCC) has raised interest in exploring agents targeting PI3K, the product of PIK3CA. The genetic and molecular basis of rigosertib treatment response was investigated in a panel of 16 HNSCC cell lines, and direct patient tumor xenografts from eight patients with HNSCC [four HPV-serotype16 (HPV16)-positive]. HNSCC cell lines and xenografts were characterized by pathway enrichment gene expression analysis, exon sequencing, gene copy number, Western blotting, and immunohistochemistry (IHC). Rigosertib had potent antiproliferative effects on 11 of 16 HPV(-) HNSCC cell lines. Treatment sensitivity was confirmed in two cell lines using an orthotopic in vivo xenograft model. Growth reduction after rigosertib treatment was observed in three of eight HNSCC direct patient tumor lines. The responsive tumor lines carried a combination of a PI3KCA-activating event (amplification or mutation) and a p53-inactivating event (either HPV16- or mutation-mediated TP53 inactivation). In this study, we evaluated the in vitro and in vivo efficacy of rigosertib in both HPV(+) and HPV(-) HNSCCs, focusing on inhibition of the PI3K pathway. Although consistent inhibition of the PI3K pathway was not evident in HNSCC, we identified a combination of PI3K/TP53 events necessary, but not sufficient, for rigosertib sensitivity.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Glicina/análogos & derivados , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Sulfonas/administración & dosificación , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/virología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Fosfatidilinositol 3-Quinasa Clase I , Glicina/administración & dosificación , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/patología , Neoplasias de Cabeza y Cuello/virología , Humanos , Mutación , Papillomaviridae/efectos de los fármacos , Infecciones por Papillomavirus/tratamiento farmacológico , Fosfatidilinositol 3-Quinasas/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Transducción de Señal/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Quinasa Tipo Polo 1
15.
Dis Model Mech ; 5(3): 342-50, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22344740

RESUMEN

We described previously a screening protocol in Drosophila melanogaster that allows us to identify small molecules that increase the killing effect of ionizing radiation in vivo in a multicellular context. The ability of this screen to identify agents that enhance the effect of radiation in human cancer models has been validated in published proof-of-concept studies. Here we describe an agent, identified by screening through two National Cancer Institute (NCI) small molecule libraries in Drosophila, that increases the effect of radiation. This agent, Bouvardin (NSC 259968), inhibits the elongation step of protein synthesis. We find that Bouvardin enhances the killing effect of X-rays in both Drosophila larvae and in human cancer cells. More detailed analysis showed that Bouvardin also increases the effect of radiation in clonogenic assays and in human cancer xenografts in mice. Finally, we present data that Bouvardin can also increase the efficacy of taxol. Regulation of translation is important to cancer biology. Current therapies target every aspect of cancer cell proliferation from growth factor signaling to cell division, with the exception of translation elongation. Our identification of Bouvardin as an enhancer of radio- and chemo-therapeutic agents suggests that targeting this niche has the potential to improve existing cancer therapies.


Asunto(s)
Drosophila melanogaster/metabolismo , Ensayos de Selección de Medicamentos Antitumorales/métodos , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia , Paclitaxel/uso terapéutico , Péptidos Cíclicos/farmacología , Radiación Ionizante , Animales , Productos Biológicos/farmacología , Productos Biológicos/uso terapéutico , Línea Celular Tumoral , Terapia Combinada , Modelos Animales de Enfermedad , Drosophila melanogaster/efectos de los fármacos , Humanos , Ratones , Mutación/genética , Paclitaxel/farmacología , Péptidos Cíclicos/uso terapéutico , Biosíntesis de Proteínas/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
16.
PLoS One ; 7(9): e44178, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22984473

RESUMEN

Prostate cancer is the most commonly diagnosed cancer, with an estimated 240,000 new cases reported annually in the United States. Due to early detection and advances in therapies, more than 90% of patients will survive 10 years post diagnosis and treatment. Radiation is a treatment option often used to treat localized disease; however, while radiation is very effective at killing tumor cells, normal tissues are damaged as well. Potential side-effects due to prostate cancer-related radiation therapy include bowel inflammation, erectile dysfunction, urethral stricture, rectal bleeding and incontinence. Currently, radiation therapy for prostate cancer does not include the administration of therapeutic agents to reduce these side effects and protect normal tissues from radiation-induced damage. In the current study, we show that the small molecular weight antioxidant, MnTE-2-PyP, protects normal tissues from radiation-induced damage in the lower abdomen in rats. Specifically, MnTE-2-PyP protected skin, prostate, and testes from radiation-induced damage. MnTE-2-PyP also protected from erectile dysfunction, a persistent problem regardless of the type of radiation techniques used because the penile neurovascular bundles lay in the peripheral zones of the prostate, where most prostate cancers reside. Based on previous studies showing that MnTE-2-PyP, in combination with radiation, further reduces subcutaneous tumor growth, we believe that MnTE-2-PyP represents an excellent radioprotectant in combination radiotherapy for cancer in general and specifically for prostate cancer.


Asunto(s)
Antioxidantes/farmacología , Metaloporfirinas/farmacología , Neoplasias de la Próstata/radioterapia , Protectores contra Radiación/farmacología , Radioterapia/efectos adversos , Animales , Antioxidantes/farmacocinética , Daño del ADN , Disfunción Eréctil/patología , Disfunción Eréctil/fisiopatología , Masculino , Metaloporfirinas/farmacocinética , Modelos Biológicos , Pene/efectos de los fármacos , Pene/patología , Pene/fisiopatología , Pene/efectos de la radiación , Neoplasias de la Próstata/fisiopatología , Radiación Ionizante , Protectores contra Radiación/farmacocinética , Ratas , Ratas Sprague-Dawley , Piel/efectos de los fármacos , Piel/patología , Piel/fisiopatología , Piel/efectos de la radiación , Testículo/efectos de los fármacos , Testículo/patología , Testículo/fisiopatología , Testículo/efectos de la radiación
17.
Dis Model Mech ; 4(4): 496-503, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21504911

RESUMEN

Combination therapy, in which two or more agents are applied, is more effective than single therapies for combating cancer. For this reason, combinations of chemotherapy with radiation are being explored in clinical trials, albeit with an empirical approach. We developed a screen to identify, from the onset, molecules that act in vivo in conjunction with radiation, using Drosophila as a model. Screens through two small molecule libraries from the NCI Developmental Therapeutics Program yielded microtubule poisons; this class of agents is known to enhance the effect of radiation in mammalian cancer models. Here we report an analysis of one microtubule depolymerizing agent, maytansinol isobutyrate (NSC292222; maytansinol), in Drosophila and in human cancer cells. We find that the effect of maytansinol is p53 dependent in Drosophila cells and human cancer cells, that maytansinol enhances the effect of radiation in both systems, and that the combinatorial effect of drug and radiation is additive. We also uncover a differential sensitivity to maytansinol between Drosophila cells and Drosophila larvae, which illustrates the value of studying cell behavior in the context of a whole organism. On the basis of these results, we propose that Drosophila might be a useful model for unbiased screens through new molecule libraries to find cancer drugs for combination therapy.


Asunto(s)
Drosophila melanogaster/efectos de los fármacos , Drosophila melanogaster/efectos de la radiación , Maitansina/análogos & derivados , Maitansina/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia , Animales , Muerte Celular/efectos de los fármacos , Terapia Combinada , Células HCT116 , Humanos , Larva/efectos de los fármacos , Larva/efectos de la radiación , Maitansina/farmacología , Mitosis/efectos de los fármacos , Mitosis/efectos de la radiación , Mutación/genética , Radiación Ionizante , Huso Acromático/efectos de los fármacos , Huso Acromático/efectos de la radiación , Análisis de Supervivencia , Proteína p53 Supresora de Tumor/metabolismo
18.
Clin Cancer Res ; 17(16): 5402-11, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21705453

RESUMEN

PURPOSE: Alterations in Smad4 signaling and its loss cause genomic instability and head and neck squamous cell carcinoma (HNSCC), suggesting that agents that target both Smad4-dependent and -independent pathways could control HNSCC. EXPERIMENTAL DESIGN: Resveratrol efficacy was evaluated against the HNSCC cells FaDu, Cal27, Det562, and Cal27-Smad4 for viability, DNA damage, cell-cycle progression, and apoptosis, as well as γ-H2AX expression, and focus formation (γ-H2AX and Brca1). Resveratrol efficacy was also examined in nude mice for FaDu xenograft growth. Xenografts were analyzed for γ-H2AX and cleaved caspase-3. RESULTS: Resveratrol (5-50 µmol/L) suppressed viability and induced DNA damage in FaDu and Cal27 cells but not in normal human epidermal keratinocytes and human foreskin fibroblasts, showing its selectivity toward HNSCC cells; however, Det562 cells were resistant to resveratrol even at 100 µmol/L. Cal27 cells stably transfected with Smad4 showed similar resveratrol effects as parental Cal27, indicating that a lack of resveratrol effect in Det562 cells was independent of Smad4 status in these cells. Furthermore, resveratrol caused S-phase arrest and apoptotic death of FaDu and Cal27 cells together with induction of Brca1 and γ-H2AX foci. Resveratrol (50 mg/kg body weight) treatment also inhibited FaDu tumor growth in nude mice, and γ-H2AX and cleaved caspase-3 were strongly increased in xenografts from resveratrol-treated mice compared with controls. CONCLUSION: Our findings for the first time showed antiproliferative, DNA damaging, and apoptotic effects of resveratrol in HNSCC cells independent of Smad4 status, both in vitro and in vivo, suggesting that more studies are needed to establish its potential usefulness against HNSCC.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Daño del ADN , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Proteína Smad4/metabolismo , Estilbenos/farmacología , Animales , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Proteína BRCA1/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Ensayo Cometa , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Histonas/metabolismo , Humanos , Immunoblotting , Inmunohistoquímica , Masculino , Ratones , Ratones Desnudos , Mutación , Resveratrol , Proteína Smad4/genética , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Clin Cancer Res ; 17(15): 5016-25, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21673064

RESUMEN

PURPOSE: We previously reported that a fibroblast growth factor (FGF) receptor (FGFR) signaling pathway drives growth of lung cancer cell lines of squamous and large cell histologies. Herein, we explored FGFR dependency in cell lines derived from the tobacco-related malignancy, head and neck squamous cell carcinoma (HNSCC). EXPERIMENTAL DESIGN: FGF and FGFR mRNA and protein expression was assessed in nine HNSCC cell lines. Dependence on secreted FGF2 for cell growth was tested with FP-1039, an FGFR1-Fc fusion protein. FGFR and epidermal growth factor receptor (EGFR) dependence was defined by sensitivity to multiple inhibitors selective for FGFRs or EGFR. RESULTS: FGF2 was expressed in eight of the nine HNSCC cell lines examined. Also, FGFR2 and FGFR3 were frequently expressed, whereas only two lines expressed FGFR1. FP-1039 inhibited growth of HNSCC cell lines expressing FGF2, identifying FGF2 as an autocrine growth factor. FGFR inhibitors selectively reduced in vitro growth and extracellular signal-regulated kinase signaling in three HNSCC cell lines, whereas three distinct lines exhibited responsiveness to both EGFR and FGFR inhibitors. Combinations of these drugs yielded additive growth inhibition. Finally, three cell lines were highly sensitive to EGFR tyrosine kinase inhibitors (TKI) with no contribution from FGFR pathways. CONCLUSIONS: FGFR signaling was dominant or codominant with EGFR in six HNSCC lines, whereas three lines exhibited little or no role for FGFRs and were highly EGFR dependent. Thus, the HNSCC cell lines can be divided into subsets defined by sensitivity to EGFR and FGFR-specific TKIs. FGFR inhibitors may represent novel therapeutics to deploy alone or in combination with EGFR inhibitors in HNSCC.


Asunto(s)
Comunicación Autocrina , Carcinoma de Células Escamosas/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Línea Celular Tumoral , Receptores ErbB/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Humanos , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Transducción de Señal , Carcinoma de Células Escamosas de Cabeza y Cuello
20.
Free Radic Biol Med ; 47(6): 722-30, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19520157

RESUMEN

Electrophilic cyclopentenone prostaglandins (cyPGs), such as 15-deoxy-Delta(12,14)-prostaglandin J(2) (15dPGJ(2)), initiate redox-based cell signaling responses including increased intracellular glutathione (GSH) synthesis. We investigated whether cyPGs facilitated GSH efflux and if members of the ATP-binding cassette (ABC) protein family mediated the efflux. Four human cell lines were treated with 1-6 microM cyPGs for 48 h. Media and cells were harvested for GSH measurements using HPLC-EC. CyPG treatment increased extracellular GSH levels two- to threefold over controls in HN4 and C38 cells and five- to sixfold in SAEC and MDA 1586 cells and was dependent on increased GSH synthesis. Our studies show that prostaglandin D(2) and its metabolites, prostaglandin J(2) and 15dPGJ(2), specifically induce GSH efflux compared to other eicosanoids. These higher extracellular GSH levels were associated with protection from tert-butylhydroperoxide. Superarray analysis of ABC transporters suggested only ABCG2 expression had a positive relationship in the four cell types compared with extracellular GSH increases after cyPG treatment. The ABCG2 substrate Hoechst 33342 inhibited extracellular GSH increase after 15dPGJ(2) treatment. We report for the first time that ABCG2 may play a role in GSH efflux in response to cyPG treatment and may link inflammatory signaling with antioxidant adaptive responses.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Glutatión/biosíntesis , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Proteínas de Neoplasias/metabolismo , Prostaglandina D2/análogos & derivados , Mucosa Respiratoria/efectos de los fármacos , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Bencimidazoles/farmacología , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , Citoprotección/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Glutatión/genética , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , Proteínas de Neoplasias/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Estrés Oxidativo/efectos de los fármacos , Prostaglandina D2/farmacología , Prostaglandina D2/uso terapéutico , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Especificidad por Sustrato , terc-Butilhidroperóxido/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA