Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Am J Cancer Res ; 13(7): 2969-2983, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37560002

RESUMEN

Globo-H (GH), a globo-series glycosphingolipid antigen that is synthesized by key enzymes ß1,3-galactosyltransferase V (ß3GalT5), fucosyltransferase (FUT) 1 and 2, is highly expressed on a variety of epithelial cancers rendering it a promising target for cancer immunotherapy. GH-targeting antibody-drug conjugate has been demonstrated an excellent tumor growth inhibition potency in animal models across multiple cancer types including Gastric cancer (GC). This study aims to further investigate the GH roles in GC. Significant correlations were observed between high mRNA expression of GH-synthetic key enzymes and worse overall survival (OS)/post-progression survival for GC patients based on the data from "Kaplan-Meier plotter" database (n=498). The level of GH expression was evaluated in clinical adenocarcinoma samples from 105 patients with GC by immunohistochemistry based on H-score. GH expression (H score ≥ 20; 33.3%) was significantly associated with a poor disease specific survival (DSS) and invasiveness in all samples with P=0.029 and P=0.013, respectively. In addition, it is also associated with shorter DSS and OS in poorly differentiated tumors with P=0.033 and P=0.045, respectively. Particularly, with patients ≥ 65 years of age, GH expression is also significantly associated with the stages (P=0.023), differentiation grade (P=0.038), and invasiveness (P=0.026) of the cancer. Sorted GC NCI-N87 cells with high level of endogenous GH showed higher proliferative activity compared with low-GH-expressing cells based on PCNA expression. Micro-western array analysis on high-GH-expressing GC cells indicated an upregulation in HER2-related signaling proteins including phospho-AKT/P38/JNK and Cyclin D1/Cyclin E1 proteins. Moreover, GH level was shown to be correlated with expression of total HER2 and caveolin-1 in GC cells. Immunoprecipitation study suggested that there are potential interactions among GH, caveolin-1, and HER2. In conclusions, GH level was significantly associated with the worse survival and disease progression in GC patients, especially in older patients. Enhanced cell proliferation activity through interactions among GH, HER2, and caveolin-1 interactions may contribute to GH induced tumor promotion signaling in GC. GH-targeting therapy may be a viable option for the treatment of GC patients.

2.
Am J Cancer Res ; 11(6): 2911-2927, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34249435

RESUMEN

Acute myeloid leukemia (AML) is a highly heterogenous and aggressive disease with a poor prognosis, necessitating further improvements in treatment therapies. Recently, several targeted therapies have become available for specific AML populations. To identify potential new therapeutic targets for AML, we analyzed published genome wide CRISPR-based screens to generate a gene essentiality dataset across a panel of 14 human AML cell lines while eliminating common essential genes through integration analysis with core fitness genes among 324 human cancer cell lines and DepMap databases. The key glutathione metabolic enzyme, glutamate-cysteine ligase catalytic subunit (GCLC), met the selection threshold. Using CRISPR knockout, GCLC was confirmed to be essential for the cell growth, survival, clonogenicity, and leukemogenesis in AML cells but was comparatively dispensable for normal hematopoietic stem and progenitor cells (HSPCs), indicating that GCLC is a potential therapeutic target for AML. In addition, we evaluated the essentiality of GCLC in solid tumors and demonstrated that GCLC represents a synthetic lethal target for ARID1A-deficient ovarian and gastric cancers.

3.
ACS Sens ; 5(2): 296-302, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-32073836

RESUMEN

While monoclonal antibodies are the fastest-growing class of therapeutic agents, we lack a method that can directly quantify the on- and off-target binding affinities of newly developed therapeutic antibodies in crude cell lysates. As a result, some therapeutic antibody candidates could have a moderate on-target binding affinity but a high off-target binding affinity, which not only gives a reduced efficacy but triggers unwanted side effects. Here, we report a single-molecule counting method that precisely quantifies antibody-bound receptors, free receptors, and unbound antibodies in crude cell lysates, termed digital receptor occupancy assay (DRO). Compared to the traditional flow cytometry-based binding assay, DRO assay enables direct and digital quantification of the three molecular species in solution without the additional antibodies for competitive binding. When characterizing the therapeutic antibody, cetuximab, using DRO assay, we found the on-target binding ratio to be 65% and the binding constant (Kd) to be 2.4 nM, while the off-target binding causes the binding constant to decrease by 0.3 nM. Other than cultured cells, the DRO assay can be performed on tumor mouse xenograft models. Thus, DRO is a simple and highly quantitative method for cell-based antibody binding analysis which can be broadly applied to screen and validate new therapeutic antibodies.


Asunto(s)
Anticuerpos/uso terapéutico , Afinidad de Anticuerpos/fisiología , Animales , Anticuerpos/farmacología , Humanos , Ratones
4.
Cancer Cell ; 34(6): 954-969.e4, 2018 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-30537515

RESUMEN

Multiple mechanisms of resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) have been identified in EGFR-mutant non-small cell lung cancer (NSCLC); however, recurrent resistance to EGFR TKIs due to the heterogeneous mechanisms underlying resistance within a single patient remains a major challenge in the clinic. Here, we report a role of nuclear protein kinase Cδ (PKCδ) as a common axis across multiple known TKI-resistance mechanisms. Specifically, we demonstrate that TKI-inactivated EGFR dimerizes with other membrane receptors implicated in TKI resistance to promote PKCδ nuclear translocation. Moreover, the level of nuclear PKCδ is associated with TKI response in patients. The combined inhibition of PKCδ and EGFR induces marked regression of resistant NSCLC tumors with EGFR mutations.


Asunto(s)
Resistencia a Antineoplásicos/efectos de los fármacos , Proteína Quinasa C-delta/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Microscopía Confocal , Persona de Mediana Edad , Terapia Molecular Dirigida/métodos , Mutación , Proteína Quinasa C-delta/metabolismo , Interferencia de ARN
5.
Nat Commun ; 9(1): 1908, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29765039

RESUMEN

Enriched PD-L1 expression in cancer stem-like cells (CSCs) contributes to CSC immune evasion. However, the mechanisms underlying PD-L1 enrichment in CSCs remain unclear. Here, we demonstrate that epithelial-mesenchymal transition (EMT) enriches PD-L1 in CSCs by the EMT/ß-catenin/STT3/PD-L1 signaling axis, in which EMT transcriptionally induces N-glycosyltransferase STT3 through ß-catenin, and subsequent STT3-dependent PD-L1 N-glycosylation stabilizes and upregulates PD-L1. The axis is also utilized by the general cancer cell population, but it has much more profound effect on CSCs as EMT induces more STT3 in CSCs than in non-CSCs. We further identify a non-canonical mesenchymal-epithelial transition (MET) activity of etoposide, which suppresses the EMT/ß-catenin/STT3/PD-L1 axis through TOP2B degradation-dependent nuclear ß-catenin reduction, leading to PD-L1 downregulation of CSCs and non-CSCs and sensitization of cancer cells to anti-Tim-3 therapy. Together, our results link MET to PD-L1 stabilization through glycosylation regulation and reveal it as a potential strategy to enhance cancer immunotherapy efficacy.


Asunto(s)
Antígeno B7-H1/inmunología , Hexosiltransferasas/inmunología , Evasión Inmune , Proteínas de la Membrana/inmunología , Neoplasias/inmunología , Células Madre Neoplásicas/inmunología , Animales , Antígeno B7-H1/genética , Línea Celular Tumoral , ADN-Topoisomerasas de Tipo II/genética , ADN-Topoisomerasas de Tipo II/inmunología , Transición Epitelial-Mesenquimal , Femenino , Regulación Neoplásica de la Expresión Génica , Hexosiltransferasas/genética , Humanos , Proteínas de la Membrana/genética , Ratones Endogámicos BALB C , Ratones Noqueados , Neoplasias/genética , Neoplasias/fisiopatología , Células Madre Neoplásicas/citología , Proteínas de Unión a Poli-ADP-Ribosa/genética , Proteínas de Unión a Poli-ADP-Ribosa/inmunología , beta Catenina/genética , beta Catenina/inmunología
6.
Cancer Cell ; 33(4): 752-769.e8, 2018 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-29606349

RESUMEN

Pancreatic ribonuclease (RNase) is a secreted enzyme critical for host defense. We discover an intrinsic RNase function, serving as a ligand for epidermal growth factor receptor (EGFR), a member of receptor tyrosine kinase (RTK), in pancreatic ductal adenocarcinoma (PDAC). The closely related bovine RNase A and human RNase 5 (angiogenin [ANG]) can trigger oncogenic transformation independently of their catalytic activities via direct association with EGFR. Notably, high plasma ANG level in PDAC patients is positively associated with response to EGFR inhibitor erlotinib treatment. These results identify a role of ANG as a serum biomarker that may be used to stratify patients for EGFR-targeted therapies, and offer insights into the ligand-receptor relationship between RNase and RTK families.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Clorhidrato de Erlotinib/farmacología , Neoplasias Pancreáticas/patología , Ribonucleasa Pancreática/sangre , Ribonucleasa Pancreática/metabolismo , Animales , Sitios de Unión , Biomarcadores/sangre , Carcinoma Ductal Pancreático/sangre , Carcinoma Ductal Pancreático/tratamiento farmacológico , Bovinos , Línea Celular Tumoral , Receptores ErbB/química , Receptores ErbB/metabolismo , Clorhidrato de Erlotinib/uso terapéutico , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Ratones , Trasplante de Neoplasias , Neoplasias Pancreáticas/sangre , Neoplasias Pancreáticas/tratamiento farmacológico , Ribonucleasa Pancreática/química , Transducción de Señal
7.
Soft Robot ; 4(4): 390-399, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29251562

RESUMEN

Collagen microparticles have recently gained more attention as viable cell confinement blocks in many biomedical research fields. Small volume and high surface area of collagen structure improve cell confinement, viability, and proliferation. Moreover, dense collagen fiber structure can protect cells from immune destruction. The ability to produce collagen microparticles in an accurate and reliable way is of upmost importance to the advancement of many biomedical researches, especially cancer research and tissue engineering. Currently, no such fabrication technique exists due to inherent fragility of collagen. Herein, we report the very first platform, pneumatically actuated soft micromold (PASMO) device, which addresses challenges in collagen microparticle production. Our new platform uses a soft micromold with a pneumatic actuator that can produce arbitrary shapes of collagen microstructures precisely from 100 µm to over 2 mm in range and can encapsulate cells inside without damaging the shape. The duplication accuracy of more than 96% in dimensions and 90% in depth has been demonstrated. The density of collagen fiber distribution is determined to be 86.57%, which is higher than that of collagen microparticles produced by other methods. We have confirmed cell viability in collagen microparticles. We also produce Matrigel™ particles as tool to develop a xenograft cancer model. The results demonstrate that Matrigel particles created by the PASMO device can reduce cell scattering for the xenograft model and the uniformity of tumors developed in mice is 12-fold improved, which can lead to an increased accuracy of cancer metastasis studies and drug screening research. These breakthroughs in the production of modular microparticles will push the boundaries of cancer research in the near future.

8.
Am J Cancer Res ; 7(3): 657-672, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28401019

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal disease characterized by a prominent desmoplastic stroma that may constrain tumor progression but also limit the access of therapeutic drugs. In this study, we explored a tumor-targeting strategy that enlists an engineered anti-angiogenic protein consisting of endostatin and cytosine deaminase linked to uracil phosphoribosyltransferase (EndoCD). This protein selectively binds to tumor vessels to compromise tumor angiogenesis and converts the non-toxic 5-fluorocytosine (5-FC) to the cytotoxic 5-fluorouracil to produce a chemotherapeutic bystander effect at the pancreatic tumor site. We found that resveratrol increased the protein stability of EndoCD through suppression of chymotrypsin-like proteinase activity and synergistically enhances EndoCD-mediated 5-FC-induced cell killing. In various PDAC mouse models, the EndoCD/5-FC/resveratrol regimen decreased intratumoral vascular density and stroma formation and enhances apoptosis in tumors cells as well as in surrounding endothelial, pancreatic stellate, and immune cells, leading to reduced tumor growth and extended survival. Thus, the EndoCD/5-FC/resveratrol combination may be an effective treatment option for PDAC.

9.
Mol Cancer Ther ; 16(7): 1246-1256, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28428442

RESUMEN

Diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma are the most prevalent B-lymphocyte neoplasms in which abnormal activation of the Bruton tyrosine kinase (BTK)-mediated B-cell receptor signaling pathway contributes to pathogenesis. Ibrutinib is an oral covalent BTK inhibitor that has shown some efficacy in both indications. To improve ibrutinib efficacy through combination therapy, we first investigated differential gene expression in parental and ibrutinib-resistant cell lines to better understand the mechanisms of resistance. Ibrutinib-resistant TMD8 cells had higher BCL2 gene expression and increased sensitivity to ABT-199, a BCL-2 inhibitor. Consistently, clinical samples from ABC-DLBCL patients who experienced poorer response to ibrutinib had higher BCL2 gene expression. We further demonstrated synergistic growth suppression by ibrutinib and ABT-199 in multiple ABC-DLBCL, GCB-DLBCL, and follicular lymphoma cell lines. The combination of both drugs also reduced colony formation, increased apoptosis, and inhibited tumor growth in a TMD8 xenograft model. A synergistic combination effect was also found in ibrutinib-resistant cells generated by either genetic mutation or drug treatment. Together, these findings suggest a potential clinical benefit from ibrutinib and ABT-199 combination therapy. Mol Cancer Ther; 16(7); 1246-56. ©2017 AACR.


Asunto(s)
Resistencia a Antineoplásicos/genética , Linfoma Folicular/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Adenina/análogos & derivados , Agammaglobulinemia Tirosina Quinasa , Animales , Protocolos de Quimioterapia Combinada Antineoplásica , Apoptosis/efectos de los fármacos , Compuestos Bicíclicos Heterocíclicos con Puentes/administración & dosificación , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Linfoma Folicular/genética , Linfoma Folicular/patología , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/patología , Ratones , Piperidinas , Inhibidores de Proteínas Quinasas/administración & dosificación , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Pirazoles/administración & dosificación , Pirimidinas/administración & dosificación , Transducción de Señal/efectos de los fármacos , Sulfonamidas/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Am J Cancer Res ; 6(11): 2489-2501, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27904766

RESUMEN

Diffuse large B cell lymphoma (DLBCL) is a heterogeneous lymphoma and the most common subtype of non-Hodgkin lymphoma, accounting for roughly 30% of newly diagnosed cases in the United States. DLBCL can be separated into the activated B cell-like (ABC) and germinal center B cell-like (GCB) subtypes, with distinct gene expression profiles, oncogenic aberrations, and clinical outcomes. ABC-DLBCL is characterized by chronically active B-cell receptor (BCR) signaling that can be modulated by Bruton's tyrosine kinase (BTK) activity. Thus, BTK serves as an attractive therapeutic target in this type of B-cell malignancy. Ibrutinib, a first-in-class, orally available covalent BTK inhibitor, has demonstrated clinical activity in several B-cell leukemias and lymphomas. A phase 1/2 clinical trial of single-agent ibrutinib in relapsed and refractory DLBCL patients revealed an overall response rate of 37% in ABC-DLBCL patients. However, responses to kinase-directed therapies are often limited by emerging resistance mechanisms that bypass the therapeutic target. Here we report the discovery of point mutations within the kinase PIM1 that reduce sensitivity to ibrutinib in ABC-DLBCL. These mutations stabilize PIM1 and affect upstream regulators and downstream targets of NF-κB signaling. The introduction of mutant PIM1 into an ABC-DLBCL cell line, TMD8, increased colony formation and decreased sensitivity to ibrutinib. In addition, ibrutinib-resistant cell lines generated by prolonged ibrutinib exposure in vitro upregulated PIM1 expression, consistent with a role for PIM1 in antagonizing ibrutinib activity. The combination of a pan-PIM inhibitor with ibrutinib synergistically inhibited proliferation in vitro and tumor growth in vivo. Together, these data provide a rationale for combining BTK and PIM1 inhibition in the treatment of ABC-DLBCL.

12.
Nat Med ; 22(2): 194-201, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26779812

RESUMEN

Poly (ADP-ribose) polymerase (PARP) inhibitors have emerged as promising therapeutics for many diseases, including cancer, in clinical trials. One PARP inhibitor, olaparib (Lynparza, AstraZeneca), was recently approved by the FDA to treat ovarian cancer with mutations in BRCA genes. BRCA1 and BRCA2 have essential roles in repairing DNA double-strand breaks, and a deficiency of BRCA proteins sensitizes cancer cells to PARP inhibition. Here we show that the receptor tyrosine kinase c-Met associates with and phosphorylates PARP1 at Tyr907 (PARP1 pTyr907 or pY907). PARP1 pY907 increases PARP1 enzymatic activity and reduces binding to a PARP inhibitor, thereby rendering cancer cells resistant to PARP inhibition. The combination of c-Met and PARP1 inhibitors synergized to suppress the growth of breast cancer cells in vitro and xenograft tumor models, and we observed similar synergistic effects in a lung cancer xenograft tumor model. These results suggest that the abundance of PARP1 pY907 may predict tumor resistance to PARP inhibitors, and that treatment with a combination of c-Met and PARP inhibitors may benefit patients whose tumors show high c-Met expression and who do not respond to PARP inhibition alone.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama , Proliferación Celular/efectos de los fármacos , Neoplasias Pulmonares , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Anilidas/farmacología , Animales , Bencimidazoles/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Crizotinib , Humanos , Técnicas In Vitro , Indoles/farmacología , Células MCF-7 , Ratones , Trasplante de Neoplasias , Fosforilación/efectos de los fármacos , Ftalazinas/farmacología , Piperazinas/farmacología , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/efectos de los fármacos , Pirazoles/farmacología , Piridinas/farmacología , Quinolinas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Am J Cancer Res ; 5(12): 3560-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26885446

RESUMEN

Drug repurposing is a therapeutic strategy that applies drugs to treat different diseases based on new therapeutic function. Carglumic acid (Carbaglu; Orphan Europe) is an orphan drug approved by the FDA for hyperammonemia. Administration of carglumic acid for treatment of hyperammonemia has few side effects and has been used for 10 years to effectively treat hyperammonemia symptoms of both adult and pediatric patients. Here, we tested the potential of carglumic acid to be repurposed as an anticancer agent and showed that carglumic acid promotes apoptosis and inhibits cancer cell growth ina wide variety of human cancers, including pancreatic ductal adenocarcinoma, triple-negative breast cancer (TNBC), hepatoma, and lung cancer. Our data from in vivo models indicates that orally taking 10% of the carglumic acid dose currently used for the treatment of hyperammonemia ise ffective to suppress the growth of pancreatic ductal adenocarcinomaand TNBC. If given intravenously, only 5% of the carglumic acid doseis needed to be effective against TNBC. These findings suggest that carglumic acid may serve as a safe and effective therapeutic to treat both TNBC and pancreatic cancer.

14.
Sci Signal ; 7(315): rs1, 2014 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-24595109

RESUMEN

Signal transduction is a dynamic process that regulates cellular functions through multiple types of biomolecular interactions, such as the interactions between proteins and between proteins and nucleic acids. However, the techniques currently available for identifying protein-protein or protein-nucleic acid complexes typically provide information about the overall population of signaling complexes in a sample instead of information about the individual signaling complexes therein. We developed a technique called "microchannel for multiparameter analysis of proteins in a single complex" (mMAPS) that simultaneously detected individual target proteins either singly or in a multicomponent complex in cell or tissue lysates. We detected the target proteins labeled with fluorophores by flow proteometry, which provided quantified data in the form of multidimensional fluorescence plots. Using mMAPS, we quantified individual complexes of epidermal growth factor (EGF) with its receptor EGFR, EGFR with signal transducer and activator of transcription 3 (STAT3), and STAT3 with the acetylase p300 and DNA in lysates from cultured cells with and without treatment with EGF, as well as in lysates from tumor xenograft tissue. Consistent with the ability of this method to reveal the dynamics of signaling protein interactions, we observed that cells treated with EGF induced the interaction of EGF with EGFR and the autophosphorylation of EGFR, but this interaction decreased with longer treatment time. Thus, we expect that this technique may reveal new aspects of molecular interaction dynamics.


Asunto(s)
Técnicas de Laboratorio Clínico/métodos , Complejos Multiproteicos/análisis , Mapas de Interacción de Proteínas/fisiología , Proteómica/métodos , Transducción de Señal/fisiología , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Fluorescencia , Complejos Multiproteicos/fisiología , Factor de Transcripción STAT3/metabolismo
15.
Cancer Cell ; 23(6): 796-810, 2013 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-23764002

RESUMEN

Epidermal growth factor receptor (EGFR) initiates a signaling cascade that leads to DNA synthesis and cell proliferation, but its role in regulating DNA replication licensing is unclear. Here, we show that activated EGFR phosphorylates the p56 isoform of Lyn, p56(Lyn), at Y32, which then phosphorylates MCM7, a licensing factor critical for DNA replication, at Y600 to increase its association with other minichromosome maintenance complex proteins, thereby promoting DNA synthesis complex assembly and cell proliferation. Both p56(Lyn) Y32 and MCM7 Y600 phosphorylation are enhanced in proliferating cells and correlated with poor survival of breast cancer patients. These results establish a signaling cascade in which EGFR enhances MCM7 phosphorylation and DNA replication through Lyn phosphorylation in human cancer cells.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas de Ciclo Celular/fisiología , Replicación del ADN/fisiología , Proteínas de Unión al ADN/fisiología , Receptores ErbB/fisiología , Proteínas Nucleares/fisiología , Familia-src Quinasas/metabolismo , Animales , Neoplasias de la Mama/patología , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Proteínas de Unión al ADN/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Femenino , Humanos , Ratones , Componente 7 del Complejo de Mantenimiento de Minicromosoma , Proteínas Nucleares/metabolismo , Fosforilación , Pronóstico , Transducción de Señal , Tirosina/química , Tirosina/metabolismo , Familia-src Quinasas/fisiología
16.
Am J Transl Res ; 5(4): 393-403, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23724163

RESUMEN

Antiangiogenesis is a promising antitumor strategy that inhibits tumor vascular formation to suppress tumor growth. Specifically, targeting VEGF has shown therapeutic benefits in many cancer types, leading to its approval as the first antiangiogenic drug by the Food and Drug Administration in the United States. It is known, however, that patients will experience unfavorable side effects as the VEGF and/or VEGF receptor signaling pathway is also required for homeostasis in normal tissues. Moreover, due to the cytostatic nature of antiangiogenic, cancer cells that are not killed by these drugs later develop an even more malignant phenotype, resulting in tumor invasion and metastasis. Although there have been many attempts to reduce drug resistance and increase therapeutic efficacy by combining antiangiogenic drugs with chemotherapy, the cumulative toxicity of antiangiogenic combinations limits their feasibility as treatments, as chronic angiogenesis inhibition typically reduces the antitumor effect of the co-administered chemotherapeutics. To overcome these problems, it is critical to explore new strategies that limit tumor resistance and side effects and also increase the exposure of chemotherapy drugs at the tumor site. Here, we review current understanding of antiangiogenic drugs and introduce a new combination strategy that links direct antiangiogenic protein and enzyme prodrug system with dual-targeting antiangiogenic and antiproliferative therapeutic effect in tumor microenvironment. This strategy has the potential to overcome these clinical hindrances and may serve as a paradigm for the next generation of antiangiogenic drugs.

17.
Clin Cancer Res ; 19(4): 845-54, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23344263

RESUMEN

PURPOSE: Results of multiple clinical trials suggest that EGF receptor (EGFR) tyrosine kinase inhibitors (TKI) exhibit negative effects on platinum-based chemotherapy in patients with lung cancer with wild-type (WT) EGFR, but the underlying molecular mechanisms are still uncertain. Studies that identify the mechanism of how TKIs negatively affect patients with WT EGFR are important for future development of effective strategies to target lung cancer. Thus, we returned to in vitro study to investigate and determine a possible explanation for this phenomenon. EXPERIMENTAL DESIGN: We investigated the effects of TKIs and cisplatin on caspase-independent cell death (CID) and the role of CID in the efficacy of each drug and the combination. Furthermore, we studied the mechanism by which EGFR signaling pathway is involved in CID. Finally, on the basis of the identified mechanism, we tested the combinational effects of cisplatin plus suberoylanilide hydroxamic acid (SAHA) or erastin on CID. RESULTS: We found that gefitinib inhibited cisplatin-induced CID but not caspase-dependent apoptotic cell death. In WT EGFR cells, gefitinib not only inhibited CID but also failed to induce apoptosis, therefore compromising the efficacy of cisplatin. Inhibition of EGFR-ERK/AKT by gefitinib activates FOXO3a, which in turn reduces reactive oxygen species (ROS) and ROS-mediated CID. To overcome this, we showed that SAHA and erastin, the inducers of ROS-mediated CID, strongly enhanced the effect of cisplatin in WT EGFR cells. CONCLUSION: TKI-mediated inhibition of CID plays an important role in the efficacy of chemotherapy. Moreover, FOXO3a is a key factor in the negative effects of TKI by eliminating cisplatin-induced ROS.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/patología , Caspasas/metabolismo , Muerte Celular/genética , Línea Celular Tumoral , Cisplatino/administración & dosificación , Resistencia a Antineoplásicos/efectos de los fármacos , Factor de Crecimiento Epidérmico/genética , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/metabolismo , Gefitinib , Humanos , Inhibidores de Proteínas Quinasas/administración & dosificación , Quinazolinas/administración & dosificación , Transducción de Señal
18.
Mol Cancer Ther ; 11(10): 2212-21, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22826466

RESUMEN

Clinical correlation studies have clearly shown that obesity is associated with breast cancer risk and patient survival. Although several potential mechanisms linking obesity and cancers have been proposed, the detailed molecular mechanism of obesity-mediated breast tumorigenesis has not yet been critically evaluated. In this study, we evaluated the effects of obesity on mammary tumor initiation and progression using mice with genetic and diet-induced obesity bearing mammary tumor xenografts and mouse mammary tumor virus-neu transgenic mice that were fed a high-fat diet. We show that obesity promoted mammary tumor growth and development in these animal models. Moreover, the expressions of TNFα, VEGF, IKKß, and mTOR are upregulated in mammary tumors of obese mice, suggesting that the IKKß/mTOR/VEGF signaling pathway is activated by TNFα in the tumors of obese mice. More importantly, inhibitors (rapamycin, bevacizumab, and aspirin) that target members of the pathway suppressed tumorigenesis and prolonged survival more effectively in obese mice than in nonobese mice. Here, we not only identified a specific signaling pathway that contributes to mammary tumorigenesis in obese mice but also a strategy for treating obesity-mediated breast cancer.


Asunto(s)
Neoplasias de la Mama/etiología , Quinasa I-kappa B/antagonistas & inhibidores , Neoplasias Mamarias Animales/etiología , Terapia Molecular Dirigida , Obesidad/complicaciones , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/patología , Proliferación Celular , Transformación Celular Neoplásica/patología , Femenino , Humanos , Quinasa I-kappa B/metabolismo , Neoplasias Mamarias Animales/irrigación sanguínea , Neoplasias Mamarias Animales/patología , Neoplasias Mamarias Animales/prevención & control , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica , Obesidad/patología , Transducción de Señal , Análisis de Supervivencia , Serina-Treonina Quinasas TOR/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Mol Cell ; 45(2): 171-84, 2012 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-22196886

RESUMEN

Proinflammatory cytokine TNFα plays critical roles in promoting malignant cell proliferation, angiogenesis, and tumor metastasis in many cancers. However, the mechanism of TNFα-mediated tumor development remains unclear. Here, we show that IKKα, an important downstream kinase of TNFα, interacts with and phosphorylates FOXA2 at S107/S111, thereby suppressing FOXA2 transactivation activity and leading to decreased NUMB expression, and further activates the downstream NOTCH pathway and promotes cell proliferation and tumorigenesis. Moreover, we found that levels of IKKα, pFOXA2 (S107/111), and activated NOTCH1 were significantly higher in hepatocellular carcinoma tumors than in normal liver tissues and that pFOXA2 (S107/111) expression was positively correlated with IKKα and activated NOTCH1 expression in tumor tissues. Therefore, dysregulation of NUMB-mediated suppression of NOTCH1 by TNFα/IKKα-associated FOXA2 inhibition likely contributes to inflammation-mediated cancer pathogenesis. Here, we report a TNFα/IKKα/FOXA2/NUMB/NOTCH1 pathway that is critical for inflammation-mediated tumorigenesis and may provide a target for clinical intervention in human cancer.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Transformación Celular Neoplásica/metabolismo , Factor Nuclear 3-beta del Hepatocito/genética , Quinasa I-kappa B/metabolismo , Neoplasias Hepáticas/metabolismo , Receptor Notch1/metabolismo , Animales , Carcinoma Hepatocelular/patología , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 3-beta del Hepatocito/metabolismo , Humanos , Neoplasias Hepáticas Experimentales/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Modelos Biológicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Fosforilación , Receptor Notch1/genética , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
20.
Mol Cancer Ther ; 10(8): 1327-36, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21610170

RESUMEN

Several antiangiogenic drugs targeting VEGF/VEGF receptor (VEGFR) that were approved by the Food and Drug Administration for many cancer types, including colorectal and lung cancer, can effectively reduce tumor growth. However, targeting the VEGF signaling pathway will probably influence the normal function of endothelial cells in maintaining homeostasis and can cause unwanted adverse effects. Indeed, emerging experimental evidence suggests that VEGF-targeting therapy induced less tumor cell-specific cytotoxicity, allowing residual cells to become more resistant and eventually develop a more malignant phenotype. We report an antitumor therapeutic EndoCD fusion protein developed by linking endostatin (Endo) to cytosine deaminase and uracil phosphoribosyltransferase (CD). Specifically, Endo possesses tumor antiangiogenesis activity that targets tumor endothelial cells, followed by CD, which converts the nontoxic prodrug 5-fluorocytosine (5-FC) to the cytotoxic antitumor drug 5-fluorouracil (5-FU) in the local tumor area. Moreover, selective targeting of tumor sites allows an increasing local intratumoral concentration of 5-FU, thus providing high levels of cytotoxic activity. We showed that treatment with EndoCD plus 5-FC, compared with bevacizumab plus 5-FU treatment, significantly increased the 5-FU concentration around tumor sites and suppressed tumor growth and metastasis in human breast and colorectal orthotropic animal models. In addition, in contrast to treatment with bevacizumab/5-FU, EndoCD/5-FC did not induce cardiotoxicity leading to heart failure in mice after long-term treatment. Our results showed that, compared with currently used antiangiogenic drugs, EndoCD possesses potent anticancer activity with virtually no toxic effects and does not increase tumor invasion or metastasis. Together, these findings suggest that EndoCD/5-FC could become an alternative option for future antiangiogenesis therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Citosina Desaminasa/uso terapéutico , Endostatinas/uso terapéutico , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Pentosiltransferasa/uso terapéutico , Animales , Antineoplásicos/toxicidad , Línea Celular Tumoral , Citosina Desaminasa/genética , Modelos Animales de Enfermedad , Endostatinas/genética , Terapia Genética , Vectores Genéticos/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Pentosiltransferasa/genética , Profármacos/metabolismo , Profármacos/farmacología , Profármacos/toxicidad , Proteínas Recombinantes de Fusión/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...