Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 110(47): 18946-51, 2013 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-24158479

RESUMEN

Smoking is a significant risk factor for lung cancer, the leading cause of cancer-related deaths worldwide. Although microRNAs are regulators of many airway gene-expression changes induced by smoking, their role in modulating changes associated with lung cancer in these cells remains unknown. Here, we use next-generation sequencing of small RNAs in the airway to identify microRNA 4423 (miR-4423) as a primate-specific microRNA associated with lung cancer and expressed primarily in mucociliary epithelium. The endogenous expression of miR-4423 increases as bronchial epithelial cells undergo differentiation into mucociliary epithelium in vitro, and its overexpression during this process causes an increase in the number of ciliated cells. Furthermore, expression of miR-4423 is reduced in most lung tumors and in cytologically normal epithelium of the mainstem bronchus of smokers with lung cancer. In addition, ectopic expression of miR-4423 in a subset of lung cancer cell lines reduces their anchorage-independent growth and significantly decreases the size of the tumors formed in a mouse xenograft model. Consistent with these phenotypes, overexpression of miR-4423 induces a differentiated-like pattern of airway epithelium gene expression and reverses the expression of many genes that are altered in lung cancer. Together, our results indicate that miR-4423 is a regulator of airway epithelium differentiation and that the abrogation of its function contributes to lung carcinogenesis.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinogénesis/metabolismo , Diferenciación Celular/fisiología , Neoplasias Pulmonares/diagnóstico , MicroARNs/metabolismo , Mucosa Respiratoria/citología , Animales , Biomarcadores de Tumor/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Inmunohistoquímica , Hibridación in Situ , Neoplasias Pulmonares/genética , Ratones , MicroARNs/genética , Análisis por Micromatrices , Reacción en Cadena en Tiempo Real de la Polimerasa , Mucosa Respiratoria/metabolismo
2.
Semin Respir Crit Care Med ; 32(1): 32-43, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21500122

RESUMEN

Lung carcinogenesis is a complex, stepwise process that involves the acquisition of genetic mutations and epigenetic changes that alter cellular processes, such as proliferation, differentiation, invasion, and metastasis. Here, we review some of the latest concepts in the pathogenesis of lung cancer and highlight the roles of inflammation, the "field of cancerization," and lung cancer stem cells in the initiation of the disease. Furthermore, we review how high throughput genomics, transcriptomics, epigenomics, and proteomics are advancing the study of lung carcinogenesis. Finally, we reflect on the potential of current in vitro and in vivo models of lung carcinogenesis to advance the field and on the areas of investigation where major breakthroughs will lead to the identification of novel chemoprevention strategies and therapies for lung cancer.


Asunto(s)
Genómica/métodos , Inflamación/complicaciones , Neoplasias Pulmonares/etiología , Animales , Anticarcinógenos/farmacología , Epigénesis Genética , Epigenómica/métodos , Perfilación de la Expresión Génica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Inflamación/fisiopatología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Mutación , Células Madre Neoplásicas/metabolismo , Proteómica/métodos
3.
Cancers (Basel) ; 13(17)2021 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-34503075

RESUMEN

CA-125, encoded by the MUC16 gene, is highly expressed in most ovarian cancer cells and thus serves as a tumor marker for monitoring disease progression or treatment response in ovarian cancer patients. However, targeting MUC16/CA-125 for ovarian cancer treatment has not been successful to date. In the current study, we performed multiple steps of high-fidelity PCR and obtained a 5 kb DNA fragment upstream of the human MUC16 gene. Reporter assays indicate that this DNA fragment possesses transactivation activity in CA-125-high cancer cells, but not in CA-125-low cancer cells, indicating that the DNA fragment contains the transactivation region that controls specific expression of the MUC16 gene in ovarian cancer cells. We further refined the promoter and found a 1040 bp fragment with similar transcriptional activity and specificity. We used this refined MUC16 promoter to replace the E1A promoter in the adenovirus type 5 genome DNA, where E1A is an essential gene for adenovirus replication. We then generated a conditionally replicative oncolytic adenovirus (CRAd) that replicates in and lyses CA-125-high cancer cells, but not CA-125-low or -negative cancer cells. In vivo studies showed that intraperitoneal virus injection prolonged the survival of NSG mice inoculated intraperitoneally (ip) with selected ovarian cancer cell lines. Furthermore, the CRAd replicates in and lyses primary ovarian cancer cells, but not normal cells, collected from ovarian cancer patients. Collectively, these data indicate that targeting MUC16 transactivation utilizing CRAd is a feasible approach for ovarian cancer treatment that warrants further investigation.

4.
Am J Transl Res ; 12(2): 409-427, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32194893

RESUMEN

Oncogenic KRAS mutations are frequently found in non-small cell lung carcinoma (NSCLC) and cause constitutive activation of the MEK-ERK pathway. Many cancer types have been shown to overexpress PD-L1 to escape immune surveillance. FRA1 is a MEK/ERK-dependent oncogenic transcription factor and a member of the AP-1 transcriptional factor superfamily. This study assesses the hypothesis that KRAS mutation directly regulates PD-L1 expression through the MEK-ERK pathway mediated by FRA1. Premalignant human bronchial epithelial cell (HBEC) lines harboring the KRAS mutationV12, EGFR mutation, p53 knock-down, or both KRAS mutation and p53 knock-down were tested for levels of PD-L1, FRA1, and ERK activation (pERK). Our results showed that KRAS mutation alone, but not other genetic alterations, induced significantly higher expression of PD-L1 compared to its vector counterparts. The increased PD-L1 expression in the KRAS mutated cells was dramatically reduced by inhibition of ERK activation. Furthermore, the MEK-ERK pathway-dependent PD-L1 expression was markedly reduced by FRA1 silencing. Interestingly, FRA1 silencing led to inhibition of ERK activation, indicating that FRA1 plays a role in PD-L1 regulation via positive feedback of ERK activation. Correlation of PD-L1 and FRA1 mRNA expression was validated using human lung cancer specimens from The Cancer Genome Atlas (TCGA) and established NSCLC cell lines from Cancer Cell Line Encyclopedia (CCLE). FRA1 expression was significantly associated with PD-L1 expression, and high FRA1 expression was correlated with poor overall survival. Our findings suggest that oncogenic KRAS-driven PD-L1 expression is dependent on MEK-ERK and FRA1 in high risk, premalignant HBEC.

6.
Sci Rep ; 10(1): 377, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31941995

RESUMEN

Chronic inflammation facilitates tumor progression. We discovered that a subset of non-small cell lung cancer cells underwent a gradually progressing epithelial-to-mesenchymal (EMT) phenotype following a 21-day exposure to IL-1ß, an abundant proinflammatory cytokine in the at-risk for lung cancer pulmonary and the lung tumor microenvironments. Pathway analysis of the gene expression profile and in vitro functional studies revealed that the EMT and EMT-associated phenotypes, including enhanced cell invasion, PD-L1 upregulation, and chemoresistance, were sustained in the absence of continuous IL-1ß exposure. We referred to this phenomenon as EMT memory. Utilizing a doxycycline-controlled SLUG expression system, we found that high expression of the transcription factor SLUG was indispensable for the establishment of EMT memory. High SLUG expression in tumors of lung cancer patients was associated with poor survival. Chemical or genetic inhibition of SLUG upregulation prevented EMT following the acute IL-1ß exposure but did not reverse EMT memory. Chromatin immunoprecipitation and methylation-specific PCR further revealed a SLUG-mediated temporal regulation of epigenetic modifications, including accumulation of H3K27, H3K9, and DNA methylation, in the CDH1 (E-cadherin) promoter following the chronic IL-1ß exposure. Chemical inhibition of DNA methylation not only restored E-cadherin expression in EMT memory, but also primed cells for chemotherapy-induced apoptosis.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Epigénesis Genética , Transición Epitelial-Mesenquimal , Memoria Inmunológica/inmunología , Inflamación/inmunología , Interleucina-1beta/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Humanos , Memoria Inmunológica/genética , Inflamación/genética , Interleucina-1beta/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Fenotipo , Células Tumorales Cultivadas
7.
Curr Opin Pulm Med ; 15(4): 303-7, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19417670

RESUMEN

PURPOSE OF REVIEW: Smoking is a major risk factor for lung cancer, which is the leading cause of cancer-related deaths both in the USA and worldwide. Chronic obstructive pulmonary disease and emphysema are comorbid conditions often found in lung cancer patients. The inflammatory pathways that link chronic obstructive pulmonary disease, emphysema, and lung cancer likely involve genetic and epigenetic modulations due to chronic tissue injury and abnormal tumor immunity in susceptible hosts. RECENT FINDINGS: Chronic airway inflammation contributes to alterations in the bronchial epithelium and lung microenvironment, provoking a milieu conducive to pulmonary carcinogenesis. For example, inflammation-inducible cyclooxygenase-2 is upregulated in nonsmall cell lung cancer and also plays an important role in promoting epithelial-to-mesenchymal transition. Genetic changes in the airway epithelium of smokers may help predict or identify individuals at risk for lung cancer. Finally, radiographic findings of emphysema have been established as independent risk factors for lung cancer. SUMMARY: The relationships between inflammation, airflow obstruction, and lung cancer are complex. Deregulated inflammation is complicit in the pathogenesis of chronic obstructive pulmonary disease and lung cancer, but the overlap of signaling events is not yet fully understood. Tobacco exposure is an important risk factor that confers long-term risk of lung disease. Diagnostic sensitivity of detecting lung cancer may improve with the utilization of genetic profiling in combination with pathologic evaluation of airway epithelium. Additional research is required to understand the role of epithelial-to-mesenchymal transition in chronic inflammatory lung diseases and lung carcinogenesis.


Asunto(s)
Neoplasias Pulmonares/epidemiología , Neumonía/complicaciones , Enfermedad Pulmonar Obstructiva Crónica/complicaciones , Enfermedad Crónica , Predisposición Genética a la Enfermedad/genética , Humanos , Neoplasias Pulmonares/inducido químicamente , Neoplasias Pulmonares/genética , Factores de Riesgo , Fumar/efectos adversos
8.
Cancer Res ; 79(19): 5022-5033, 2019 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-31142513

RESUMEN

Epithelial cells in the field of lung injury can give rise to distinct premalignant lesions that may bear unique genetic aberrations. A subset of these lesions may escape immune surveillance and progress to invasive cancer; however, the mutational landscape that may predict progression has not been determined. Knowledge of premalignant lesion composition and the associated microenvironment is critical for understanding tumorigenesis and the development of effective preventive and interception strategies. To identify somatic mutations and the extent of immune cell infiltration in adenomatous premalignancy and associated lung adenocarcinomas, we sequenced exomes from 41 lung cancer resection specimens, including 89 premalignant atypical adenomatous hyperplasia lesions, 15 adenocarcinomas in situ, and 55 invasive adenocarcinomas and their adjacent normal lung tissues. We defined nonsynonymous somatic mutations occurring in both premalignancy and the associated tumor as progression-associated mutations whose predicted neoantigens were highly correlated with infiltration of CD8+ and CD4+ T cells as well as upregulation of PD-L1 in premalignant lesions, suggesting the presence of an adaptive immune response to these neoantigens. Each patient had a unique repertoire of somatic mutations and associated neoantigens. Collectively, these results provide evidence for mutational heterogeneity, pathway dysregulation, and immune recognition in pulmonary premalignancy.Significance: These findings identify progression-associated somatic mutations, oncogenic pathways, and association between the mutational landscape and adaptive immune responses in adenomatous premalignancy.See related commentary by Merrick, p. 4811.


Asunto(s)
Adenocarcinoma , Adenoma , Neoplasias Pulmonares , Lesiones Precancerosas , Genómica , Humanos , Microambiente Tumoral
9.
Cancer Res ; 66(15): 7701-7, 2006 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-16885372

RESUMEN

Tumor cells aberrantly express chemokines and/or chemokine receptors, and some may promote tumor growth and metastasis. We examined the expression and function of chemokine receptor CXCR3 in a syngeneic murine model of metastatic breast cancer. By flow cytometry, CXCR3 was detected in all murine mammary tumor cell lines examined. All human breast cancer cell lines examined also expressed CXCR3, as did the immortalized but nontumorigenic MCF-10A cell line. Interaction of CXCR3 ligands, CXCL9, CXCL10, and CXCL11, with CXCR3 on the highly malignant murine mammary tumor cell line 66.1 resulted in intracellular calcium mobilization and chemotaxis in vitro. To test the hypothesis that tumor metastasis is facilitated by CXCR3 expressed by tumor cells, we employed a small molecular weight antagonist of CXCR3, AMG487. 66.1 tumor cells were pretreated with AMG487 prior to i.v. injection into immune-competent female mice. Antagonism of CXCR3 on 66.1 tumor cells inhibited experimental lung metastasis, and this antimetastatic activity was compromised in mice depleted of natural killer cells. Systemic administration of AMG487 also inhibited experimental lung metastasis. In contrast to the antimetastatic effect of AMG487, local growth of 66.1 mammary tumors was not affected by receptor antagonism. These studies indicate that murine mammary tumor cells express CXCR3 which facilitates the development of lung metastases. These studies also indicate for the first time that a small molecular weight antagonist of CXCR3 has the potential to inhibit tumor metastasis.


Asunto(s)
Neoplasias de la Mama/patología , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Receptores de Quimiocina/antagonistas & inhibidores , Adenocarcinoma/inmunología , Adenocarcinoma/prevención & control , Adenocarcinoma/secundario , Animales , Neoplasias de la Mama/inmunología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones SCID , Receptores CXCR3 , Receptores de Quimiocina/biosíntesis , Receptores de Quimiocina/inmunología
10.
Cancer Res ; 78(8): 1986-1999, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29431637

RESUMEN

Epithelial-to-mesenchymal transition (EMT) is organized in cancer cells by a set of key transcription factors, but the significance of this process is still debated, including in non-small cell lung cancer (NSCLC). Here, we report increased expression of the EMT-inducing transcription factor Snail in premalignant pulmonary lesions, relative to histologically normal pulmonary epithelium. In immortalized human pulmonary epithelial cells and isogenic derivatives, we documented Snail-dependent anchorage-independent growth in vitro and primary tumor growth and metastatic behavior in vivo Snail-mediated transformation relied upon silencing of the tumor-suppressive RNA splicing regulatory protein ESRP1. In clinical specimens of NSCLC, ESRP1 loss was documented in Snail-expressing premalignant pulmonary lesions. Mechanistic investigations showed that Snail drives malignant progression in an ALDH+CD44+CD24- pulmonary stem cell subset in which ESRP1 and stemness-repressing microRNAs are inhibited. Collectively, our results show how ESRP1 loss is a critical event in lung carcinogenesis, and they identify new candidate directions for targeted therapy of NSCLC.Significance: This study defines a Snail-ESRP1 cancer axis that is crucial for human lung carcinogenesis, with implications for new intervention strategies and translational opportunities. Cancer Res; 78(8); 1986-99. ©2018 AACR.


Asunto(s)
Transformación Celular Neoplásica/genética , Silenciador del Gen , Pulmón/patología , Proteínas de Unión al ARN/genética , Factores de Transcripción de la Familia Snail/fisiología , Animales , Línea Celular Transformada , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Pulmón/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Modelos Animales
11.
Cancer Cell ; 33(5): 905-921.e5, 2018 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-29763624

RESUMEN

Altered metabolism is a hallmark of cancer growth, forming the conceptual basis for development of metabolic therapies as cancer treatments. We performed in vivo metabolic profiling and molecular analysis of lung squamous cell carcinoma (SCC) to identify metabolic nodes for therapeutic targeting. Lung SCCs adapt to chronic mTOR inhibition and suppression of glycolysis through the GSK3α/ß signaling pathway, which upregulates glutaminolysis. Phospho-GSK3α/ß protein levels are predictive of response to single-therapy mTOR inhibition while combinatorial treatment with the glutaminase inhibitor CB-839 effectively overcomes therapy resistance. In addition, we identified a conserved metabolic signature in a broad spectrum of hypermetabolic human tumors that may be predictive of patient outcome and response to combined metabolic therapies targeting mTOR and glutaminase.


Asunto(s)
Bencenoacetamidas/administración & dosificación , Compuestos de Boro/administración & dosificación , Carcinoma de Células Escamosas/metabolismo , Glutamina/metabolismo , Glicina/análogos & derivados , Glucógeno Sintasa Quinasa 3/metabolismo , Neoplasias Pulmonares/metabolismo , Tiadiazoles/administración & dosificación , Animales , Bencenoacetamidas/farmacología , Compuestos de Boro/farmacología , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glicina/administración & dosificación , Glicina/farmacología , Glucólisis , Humanos , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Ratones , Trasplante de Neoplasias , Transducción de Señal/efectos de los fármacos , Tiadiazoles/farmacología
12.
Cancer Prev Res (Phila) ; 10(9): 514-524, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28754664

RESUMEN

Lung cancers are documented to have remarkable intratumoral genetic heterogeneity. However, little is known about the heterogeneity of biophysical properties, such as cell motility, and its relationship to early disease pathogenesis and micrometastatic dissemination. In this study, we identified and selected a subpopulation of highly migratory premalignant airway epithelial cells that were observed to migrate through microscale constrictions at up to 100-fold the rate of the unselected immortalized epithelial cell lines. This enhanced migratory capacity was found to be Rac1-dependent and heritable, as evidenced by maintenance of the phenotype through multiple cell divisions continuing more than 8 weeks after selection. The morphology of this lung epithelial subpopulation was characterized by increased cell protrusion intensity. In a murine model of micrometastatic seeding and pulmonary colonization, the motility-selected premalignant cells exhibit both enhanced survival in short-term assays and enhanced outgrowth of premalignant lesions in longer-term assays, thus overcoming important aspects of "metastatic inefficiency." Overall, our findings indicate that among immortalized premalignant airway epithelial cell lines, subpopulations with heritable motility-related biophysical properties exist, and these may explain micrometastatic seeding occurring early in the pathogenesis of lung cancer. Understanding, targeting, and preventing these critical biophysical traits and their underlying molecular mechanisms may provide a new approach to prevent metastatic behavior. Cancer Prev Res; 10(9); 514-24. ©2017 AACRSee related editorial by Hynds and Janes, p. 491.


Asunto(s)
Bronquios/citología , Movimiento Celular/genética , Proliferación Celular/genética , Células Epiteliales/patología , Neoplasias Pulmonares/genética , Animales , Bronquios/patología , Línea Celular Tumoral , Supervivencia Celular/genética , Células Epiteliales/citología , Células Epiteliales/metabolismo , Perfilación de la Expresión Génica , Humanos , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos NOD , Organismos Libres de Patógenos Específicos , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína de Unión al GTP rac1/metabolismo
13.
Clin Cancer Res ; 23(16): 4556-4568, 2017 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-28468947

RESUMEN

Purpose: A phase I study was conducted to determine safety, clinical efficacy, and antitumor immune responses in patients with advanced non-small cell lung carcinoma (NSCLC) following intratumoral administration of autologous dendritic cells (DC) transduced with an adenoviral (Ad) vector expressing the CCL21 gene (Ad-CCL21-DC). We evaluated safety and tumor antigen-specific immune responses following in situ vaccination (ClinicalTrials.gov: NCT01574222).Experimental Design: Sixteen stage IIIB/IV NSCLC subjects received two vaccinations (1 × 106, 5 × 106, 1 × 107, or 3 × 107 DCs/injection) by CT- or bronchoscopic-guided intratumoral injections (days 0 and 7). Immune responses were assessed by tumor antigen-specific peripheral blood lymphocyte induction of IFNγ in ELISPOT assays. Tumor biopsies were evaluated for CD8+ T cells by IHC and for PD-L1 expression by IHC and real-time PCR (RT-PCR).Results: Twenty-five percent (4/16) of patients had stable disease at day 56. Median survival was 3.9 months. ELISPOT assays revealed 6 of 16 patients had systemic responses against tumor-associated antigens (TAA). Tumor CD8+ T-cell infiltration was induced in 54% of subjects (7/13; 3.4-fold average increase in the number of CD8+ T cells per mm2). Patients with increased CD8+ T cells following vaccination showed significantly increased PD-L1 mRNA expression.Conclusions: Intratumoral vaccination with Ad-CCL21-DC resulted in (i) induction of systemic tumor antigen-specific immune responses; (ii) enhanced tumor CD8+ T-cell infiltration; and (iii) increased tumor PD-L1 expression. Future studies will evaluate the role of combination therapies with PD-1/PD-L1 checkpoint inhibition combined with DC-CCL21 in situ vaccination. Clin Cancer Res; 23(16); 4556-68. ©2017 AACR.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Quimiocina CCL21/inmunología , Células Dendríticas/inmunología , Inmunoterapia Adoptiva/métodos , Neoplasias Pulmonares/terapia , Adulto , Anciano , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Antígeno B7-H1/metabolismo , Linfocitos T CD8-positivos/metabolismo , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Quimiocina CCL21/genética , Estudios de Cohortes , Células Dendríticas/metabolismo , Células Dendríticas/trasplante , Disnea/etiología , Femenino , Humanos , Inmunoterapia Adoptiva/efectos adversos , Inyecciones Intralesiones , Interferón gamma/inmunología , Interferón gamma/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Masculino , Persona de Mediana Edad , Debilidad Muscular/etiología , Dolor/etiología
14.
Cell Rep ; 18(3): 601-610, 2017 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-28099841

RESUMEN

Cancer cells exhibit increased use of nutrients, including glucose and glutamine, to support the bioenergetic and biosynthetic demands of proliferation. We tested the small-molecule inhibitor of glutaminase CB-839 in combination with erlotinib on epidermal growth factor receptor (EGFR) mutant non-small cell lung cancer (NSCLC) as a therapeutic strategy to simultaneously impair cancer glucose and glutamine utilization and thereby suppress tumor growth. Here, we show that CB-839 cooperates with erlotinib to drive energetic stress and activate the AMP-activated protein kinase (AMPK) pathway in EGFR (del19) lung tumors. Tumor cells undergo metabolic crisis and cell death, resulting in rapid tumor regression in vivo in mouse NSCLC xenografts. Consistently, positron emission tomography (PET) imaging with 18F-fluoro-2-deoxyglucose (18F-FDG) and 11C-glutamine (11C-Gln) of xenografts indicated reduced glucose and glutamine uptake in tumors following treatment with CB-839 + erlotinib. Therefore, PET imaging with 18F-FDG and 11C-Gln tracers can be used to non-invasively measure metabolic response to CB-839 and erlotinib combination therapy.


Asunto(s)
Apoptosis/efectos de los fármacos , Bencenoacetamidas/toxicidad , Receptores ErbB/antagonistas & inhibidores , Clorhidrato de Erlotinib/toxicidad , Glutaminasa/antagonistas & inhibidores , Tiadiazoles/toxicidad , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Autofagia/efectos de los fármacos , Bencenoacetamidas/uso terapéutico , Radioisótopos de Carbono/química , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico por imagen , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Receptores ErbB/genética , Receptores ErbB/metabolismo , Clorhidrato de Erlotinib/uso terapéutico , Fluorodesoxiglucosa F18/química , Glutaminasa/metabolismo , Glutamina/química , Glutamina/metabolismo , Humanos , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Ratones , Ratones SCID , Mutación , Interferencia de ARN , Radiofármacos/química , Tiadiazoles/uso terapéutico , Trasplante Heterólogo
15.
Cancer Res ; 75(22): 4910-22, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26574479

RESUMEN

Inactivation of the LKB1 tumor suppressor is a frequent event in non-small cell lung carcinoma (NSCLC) leading to the activation of mTOR complex 1 (mTORC1) and sensitivity to the metabolic stress inducer phenformin. In this study, we explored the combinatorial use of phenformin with the mTOR catalytic kinase inhibitor MLN0128 as a treatment strategy for NSCLC bearing comutations in the LKB1 and KRAS genes. NSCLC is a genetically and pathologically heterogeneous disease, giving rise to lung tumors of varying histologies that include adenocarcinomas and squamous cell carcinomas (SCC). We demonstrate that phenformin in combination with MLN0128 induced a significant therapeutic response in KRAS/LKB1-mutant human cell lines and genetically engineered mouse models of NSCLC that develop both adenocarcinomas and SCCs. Specifically, we found that KRAS/LKB1-mutant lung adenocarcinomas responded strongly to phenformin + MLN0128 treatment, but the response of SCCs to single or combined treatment with MLN0128 was more attenuated due to acquired resistance to mTOR inhibition through modulation of the AKT-GSK signaling axis. Combinatorial use of the mTOR inhibitor and AKT inhibitor MK2206 robustly inhibited the growth and viability of squamous lung tumors, thus providing an effective strategy to overcome resistance. Taken together, our findings define new personalized therapeutic strategies that may be rapidly translated into clinical use for the treatment of KRAS/LKB1-mutant adenocarcinomas and squamous cell tumors.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/genética , Estrés Fisiológico/efectos de los fármacos , Quinasas de la Proteína-Quinasa Activada por el AMP , Proteínas Quinasas Activadas por AMP , Animales , Benzoxazoles/farmacología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Inmunohistoquímica , Ratones , Fenformina/farmacología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Pirimidinas/farmacología
16.
Breast Dis ; 20: 137-43, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15687714

RESUMEN

Breast cancers, like other malignancies, commonly express a repertoire of both chemokines and chemokine receptors. While some are more often expressed in certain histological types, a few general concepts are emerging that provide clues to the pathobiological role of these ligand receptor pairs. The receptor CXCR4 is often expressed in solid tumors and evidence is growing that this receptor plays a role in the growth and lymph node metastasis of breast and other cancers responding to ligand expressed at metastatic sites. Likewise, CCR7 is expressed in breast and other cancers and, in some cases, is associated with more aggressive disease. Like chemokine receptors, some ligands also modulate tumor behavior. CCL5 expression is associated with more aggressive breast cancers. CXC chemokines containing the ELR motif are expressed endogenously by some cancers, act as autocrine growth factors and support tumor angiogenesis. ELR-negative CXC chemokines inhibit tumor growth and metastasis when expressed at high levels by attracting immune effector cells and inhibiting angiogenesis. The roles of other chemokine receptors and ligands are under active investigation.


Asunto(s)
Neoplasias de la Mama/inmunología , Neoplasias de la Mama/terapia , Quimiocinas/inmunología , Inmunoterapia , Animales , Humanos
17.
Cancer Prev Res (Phila) ; 7(1): 150-60, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24253315

RESUMEN

Definition of the molecular pathogenesis of lung cancer allows investigators an enhanced understanding of the natural history of the disease, thus fostering development of new prevention strategies. In addition to regulating epithelial-to-mesenchymal transition (EMT), the transcription factor Snail exerts global effects on gene expression. Our recent studies reveal that Snail is upregulated in non-small cell lung cancer (NSCLC), is associated with poor prognosis, and promotes tumor progression in vivo. Herein, we demonstrate that overexpression of Snail leads to the upregulation of secreted protein, acidic and rich in cysteine (SPARC) in models of premalignancy and established disease, as well as in lung carcinoma tissues in situ. Snail overexpression leads to increased SPARC-dependent invasion in vitro, indicating that SPARC may play a role in lung cancer progression. Bioinformatic analysis implicates transforming growth factor beta (TGF-ß), extracellular signal-regulated kinase (ERK)1/2, and miR-29b as potential intermediaries in Snail-mediated upregulation of SPARC. Both the TGF-ß1 ligand and TGF-ß receptor 2 (TGF-ßR2) are upregulated following Snail overexpression. Treatment of human bronchial epithelial cell (HBEC) lines with TGF-ß1 and inhibition of TGF-ß1 mRNA expression modulates SPARC expression. Inhibition of MAP-ERK kinase (MEK) phosphorylation downregulates SPARC. MiR-29b is downregulated in Snail-overexpressing cell lines, whereas overexpression of miR-29b inhibits SPARC expression. In addition, miR-29b is upregulated following ERK inhibition, suggesting a Snail-dependent pathway by which Snail activation of TGF-ß and ERK signaling results in downregulation of miR-29b and subsequent upregulation of SPARC. Our discovery of pathways responsible for Snail-induced SPARC expression contributes to the definition of NSCLC pathogenesis.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/metabolismo , Osteonectina/metabolismo , Factores de Transcripción/metabolismo , Bronquios/citología , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Progresión de la Enfermedad , Células Epiteliales/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Ligandos , Neoplasias Pulmonares/patología , MicroARNs/metabolismo , Invasividad Neoplásica , Fosforilación , Pronóstico , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal , Factores de Transcripción de la Familia Snail , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba
18.
J Cancer Sci Ther ; 6(11): 468-477, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26523208

RESUMEN

BACKGROUND: The cyclooxygenase 2 (COX-2) pathway has been implicated in the molecular pathogenesis of many malignancies, including lung cancer. Apricoxib, a selective COX-2 inhibitor, has been described to inhibit epithelial-mesenchymal transition (EMT) in human malignancies. The mechanism by which apricoxib may alter the tumor microenvironment by affecting EMT through other important signaling pathways is poorly defined. IL-27 has been shown to have anti-tumor activity and our recent study showed that IL-27 inhibited EMT through a STAT1 dominant pathway. OBJECTIVE: The purpose of this study is to investigate the role of apricoxib combined with IL-27 in inhibiting lung carcinogenesis by modulation of EMT through STAT signaling. METHODS AND RESULTS: Western blot analysis revealed that IL-27 stimulation of human non-small cell lung cancer (NSCLC) cell lines results in STAT1 and STAT3 activation, decreased Snail protein and mesenchymal markers (N-cadherin and vimentin) and a concomitant increase in expression of epithelial markers (E-cadherin, ß-and γ-catenins), and inhibition of cell migration. The combination of apricoxib and IL-27 resulted in augmentation of STAT1 activation. However, IL-27 mediated STAT3 activation was decreased by the addition of apricoxib. STAT1 siRNA was used to determine the involvement of STAT1 pathway in the enhanced inhibition of EMT and cell migration by the combined IL-27 and apricoxib treatment. Pretreatment of cells with STAT1 siRNA inhibited the effect of combined IL-27 and apricoxib in the activation of STAT1 and STAT3. In addition, the augmented expression of epithelial markers, decreased expression mesenchymal markers, and inhibited cell migration by the combination treatment were also inhibited by STAT1 siRNA, suggesting that the STAT1 pathway is important in the enhanced effect from the combination treatment. CONCLUSION: Combined apricoxib and IL-27 has an enhanced effect in inhibition of epithelial-mesenchymal transition and cell migration in human lung cancer cells through a STAT1 dominant pathway.

19.
Cancer Prev Res (Phila) ; 7(8): 845-55, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24913817

RESUMEN

Understanding the molecular pathogenesis of lung cancer is necessary to identify biomarkers/targets specific to individual airway molecular profiles and to identify options for targeted chemoprevention. Herein, we identify mechanisms by which loss of microRNA (miRNA)125a-3p (miR125a) contributes to the malignant potential of human bronchial epithelial cells (HBEC) harboring an activating point mutation of the K-ras proto-oncogene (HBEC K-ras). Among other miRNAs, we identified significant miR125a loss in HBEC K-ras lines and determined that miR125a is regulated by the PEA3 transcription factor. PEA3 is upregulated in HBEC K-ras cells, and genetic knockdown of PEA3 restores miR125a expression. From a panel of inflammatory/angiogenic factors, we identified increased CXCL1 and vascular endothelial growth factor (VEGF) production by HBEC K-ras cells and determined that miR125a overexpression significantly reduces K-ras-mediated production of these tumorigenic factors. miR125a overexpression also abrogates increased proliferation of HBEC K-ras cells and suppresses anchorage-independent growth (AIG) of HBEC K-ras/P53 cells, the latter of which is CXCL1-dependent. Finally, pioglitazone increases levels of miR125a in HBEC K-ras cells via PEA3 downregulation. In addition, pioglitazone and miR125a overexpression elicit similar phenotypic responses, including suppression of both proliferation and VEGF production. Our findings implicate miR125a loss in lung carcinogenesis and lay the groundwork for future studies to determine whether miR125a is a possible biomarker for lung carcinogenesis and/or a chemoprevention target. Moreover, our studies illustrate that pharmacologic augmentation of miR125a in K-ras-mutated pulmonary epithelium effectively abrogates several deleterious downstream events associated with the mutation.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Genes ras , Neoplasias Pulmonares/metabolismo , MicroARNs/metabolismo , Factores de Transcripción/metabolismo , Bronquios/citología , Línea Celular , Proliferación Celular , Quimiocina CXCL1/metabolismo , Ensayo de Inmunoadsorción Enzimática , Epitelio/metabolismo , Humanos , Neoplasias Pulmonares/genética , Mutación , Pioglitazona , Mutación Puntual , Lesiones Precancerosas/metabolismo , Proto-Oncogenes Mas , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Tiazolidinedionas/química , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas ras/metabolismo
20.
Mol Cancer Res ; 12(5): 765-74, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24469837

RESUMEN

UNLABELLED: Aberrant expression of microRNAs (miRNA) with oncogenic capacities (oncomiRs) has been described for several different malignancies. The first identified oncomiR, miR-17-92, is frequently overexpressed in a variety of cancers and its targets include the tumor suppressor PTEN. The transcription factor c-Myc (MYC) plays a central role in proliferative control and is rapidly upregulated upon mitogenic stimulation. Expression of c-Myc is frequently deregulated in tumors, facilitating proliferation and inhibiting terminal differentiation. The c-Myc-regulated network comprises a large number of transcripts, including those encoding miRNAs. Here, prostaglandin E2 (PGE2) exposure rapidly upregulates the expression of the MYC gene followed by the elevation of miR-17-92 levels, which in turn suppresses PTEN expression, thus enhancing apoptosis resistance in non-small cell lung cancer (NSCLC) cells. Knockdown of MYC expression or the miR-17-92 cluster effectively reverses this outcome. Similarly, miR-17-92 levels are significantly elevated in NSCLC cells ectopically expressing COX-2. Importantly, circulating miR-17-92 was elevated in the blood of patients with lung cancer as compared with subjects at risk for developing lung cancer. Furthermore, in patients treated with celecoxib, miR-17-92 levels were significantly reduced. These data demonstrate that PGE2, abundantly produced by NSCLC and inflammatory cells in the tumor microenvironment, is able to stimulate cell proliferation and promote resistance to pharmacologically induced apoptosis in a c-Myc and miR-17-92-dependent manner. IMPLICATIONS: This study describes a novel mechanism, involving c-Myc and miR-17-92, which integrates cell proliferation and apoptosis resistance.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Dinoprostona/farmacología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , MicroARNs/biosíntesis , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Carcinoma de Pulmón de Células no Pequeñas/patología , Celecoxib , Procesos de Crecimiento Celular/efectos de los fármacos , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Ciclooxigenasa 2/biosíntesis , Ciclooxigenasa 2/metabolismo , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Genes Supresores de Tumor , Genes myc , Humanos , Neoplasias Pulmonares/patología , MicroARNs/sangre , MicroARNs/genética , Fosfohidrolasa PTEN/biosíntesis , Fosfohidrolasa PTEN/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Pirazoles/farmacología , ARN Largo no Codificante , Sulfonamidas/farmacología , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA