Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros












Intervalo de año de publicación
1.
Braz. J. Pharm. Sci. (Online) ; 58: e20343, 2022. tab, graf
Artículo en Inglés | LILACS | ID: biblio-1420499

RESUMEN

Abstract To evaluate possible new drugs for the treatment of Colorectal Carcinoma (CRC) using in silico tools was the main objective of this study. The method of analysis used was the in silico evaluation of tumor markers and their interaction with selected drugs, through the study of its pharmacokinetic and pharmacodynamic characteristics. A potential therapeutic target pointed out in this study was the Cell Division Cycle 25 B (CDC25B), belonging to the CDC25 phosphatase family. Overexpression of CDC25 phosphatases is often associated with a wide variety of cancers. In addition, CDC25B is an oncogenic protein that induces neoplastic transformation. In CRC, CDC25B is overexpressed to activate the CDC2/cyclin B complex and improve the growth and survival of these tumors. Four drugs were identified for evaluation, with α-amyrin being selected for docking, because it was that had the best characteristics according to the methodology used. The α-amyrin ligand obtained the interaction energy value of -7.6 G (Kcal/mol), while the standard CDC25B ligand obtained -10.0 G (Kcal/mol). TThe results showed that the CDC25B protein was the only structure cocrystallized with α-amyrin and presented favorable outcomes in docking, being a candidate for further studies for its use in the CRC targeted therapy


Asunto(s)
Simulación por Computador , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Biomarcadores de Tumor/agonistas
2.
Int Immunopharmacol ; 97: 107681, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33932697

RESUMEN

Cancer is a leading cause of death worldwide and imposes a substantial financial burden. Therefore, it is essential to develop cost-effective approaches to inhibit tumor growth and development. The imbalance of cytokines and chemokines play an important role among different mechanisms involved in cancer development. One of the strongly conserved chemokines that is constitutively expressed in skin epithelia is the chemokine CXCL14. As a member of the CXC subfamily of chemokines, CXCL14 is responsible for the infiltration of immune cells, maturation of dendritic cells, upregulation of major histocompatibility complex (MHC)-I expression, and cell mobilization. Moreover, dysregulation of CXCL14 in several cancers has been identified by several studies. Depending on the type or origin of the tumor and components of the tumor microenvironment, CXCL14 plays a conflicting role in cancer. Although fibroblast-derived CXCL14 has a tumor-supportive role, epithelial-derived CXCL14 mainly inhibits tumor progression. Hence, this review will elucidate what is known on the mechanisms of CXCL14 and its therapeutic approaches in tumor treatment. CXCL14 is a promising approach for cancer immunotherapy.


Asunto(s)
Antineoplásicos/farmacología , Biomarcadores de Tumor/metabolismo , Quimiocinas CXC/metabolismo , Neoplasias/inmunología , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/agonistas , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/antagonistas & inhibidores , Quimiocinas CXC/agonistas , Quimiocinas CXC/análisis , Quimiocinas CXC/antagonistas & inhibidores , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/inmunología , Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Regulación hacia Arriba
3.
Theranostics ; 11(7): 3262-3277, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33537086

RESUMEN

Semaphorins are a large family of developmental regulatory signals, characterized by aberrant expression in human cancers. These molecules crucially control cell-cell communication, cell migration, invasion and metastasis, tumor angiogenesis, inflammatory and anti-cancer immune responses. Semaphorins comprise secreted and cell surface-exposed molecules and their receptors are mainly found in the Plexin and Neuropilin families, which are further implicated in a signaling network controlling the tumor microenvironment. Accumulating evidence indicates that semaphorins may be considered as novel clinical biomarkers for cancer, especially for the prediction of patient survival and responsiveness to therapy. Moreover, preclinical experimental studies have demonstrated that targeting semaphorin signaling can interfere with tumor growth and/or metastatic dissemination, suggesting their relevance as novel therapeutic targets in cancer; this has also prompted the development of semaphorin-interfering molecules for application in the clinic. Here we will survey, in diverse human cancers, the current knowledge about the relevance of semaphorin family members, and conceptualize potential lines of future research development in this field.


Asunto(s)
Biomarcadores de Tumor/genética , Moléculas de Adhesión Celular/genética , Neoplasias/genética , Neovascularización Patológica/genética , Proteínas del Tejido Nervioso/genética , Neuropilinas/genética , Semaforinas/genética , Animales , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/agonistas , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/metabolismo , Moléculas de Adhesión Celular/metabolismo , Comunicación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Humanos , Metástasis de la Neoplasia , Neoplasias/tratamiento farmacológico , Neoplasias/mortalidad , Neoplasias/patología , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/mortalidad , Neovascularización Patológica/patología , Proteínas del Tejido Nervioso/metabolismo , Neuropilinas/metabolismo , Pronóstico , Semaforinas/agonistas , Semaforinas/antagonistas & inhibidores , Semaforinas/metabolismo , Transducción de Señal , Análisis de Supervivencia , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética
5.
Discov Med ; 29(156): 27-39, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32598860

RESUMEN

The dysfunction of long non-coding RNAs (lncRNAs), without protein-coding potential, has been implicated in drug resistance against treatment in various human diseases, especially in malignant tumors. As the most common-diagnosed female malignancy worldwide, breast cancer is also the second-leading cause of cancer-related mortality in women. Despite the improvement in neo-adjuvant therapy, endocrine therapy, molecular-targeted treatment, and chemotherapy, drug resistance to various treatment regimens is still quite prevalent. This article focused on the lncRNAs and their functions in drug resistance against breast cancer therapeutic agents, in order to develop new precise treatment strategies for patients with breast cancers. The discovery of lncRNA opened new doors to the molecular mechanisms of the biological processes, and has provided new pathways to regulate biochemical events. Thus, lncRNAs may be developed as a biomarker for the detection and/or prevention of breast cancer. Additionally, lncRNA-based approaches may provide an additional treatment modality in personalized medicine alone or in combination with existing tumor-directed interventions to improve patient outcomes. In conclusion, lncRNAs molecules may represent the "next generation" therapy option for breast cancer patients.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/terapia , Resistencia a Antineoplásicos/genética , ARN Largo no Codificante/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/agonistas , Biomarcadores de Tumor/antagonistas & inhibidores , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/genética , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Quimioradioterapia/métodos , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Medicina de Precisión/métodos , ARN Largo no Codificante/agonistas , ARN Largo no Codificante/antagonistas & inhibidores , Tolerancia a Radiación/efectos de los fármacos , Tolerancia a Radiación/genética
6.
Biochim Biophys Acta Rev Cancer ; 1873(2): 188362, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32298747

RESUMEN

Trefoil factors 1, 2, and 3 (TFFs) are a family of small secretory molecules involved in the protection and repair of the gastrointestinal tract (GI). TFFs maintain and restore epithelial structural integrity via transducing key signaling pathways for epithelial cell migration, proliferation, and invasion. In recent years, TFFs have emerged as key players in the pathogenesis of multiple diseases, especially cancer. Initially recognized as tumor suppressors, emerging evidence demonstrates their key role in tumor progression and metastasis, extending their actions beyond protection. However, to date, a comprehensive understanding of TFFs' mechanism of action in tumor initiation, progression and metastasis remains obscure. The present review discusses the structural, functional and mechanistic implications of all three TFF family members in tumor progression and metastasis. Also, we have garnered information from studies on their structure and expression status in different organs, along with lessons from their specific knockout in mouse models. In addition, we highlight the emerging potential of using TFFs as a biomarker to stratify tumors for better therapeutic intervention.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias/patología , Proteínas Oncogénicas/metabolismo , Factores Trefoil/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/agonistas , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/antagonistas & inhibidores , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Epigénesis Genética/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Membrana Mucosa/metabolismo , Estadificación de Neoplasias , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/mortalidad , Proteínas Oncogénicas/análisis , Proteínas Oncogénicas/antagonistas & inhibidores , Pronóstico , Dominios Proteicos , Factores Trefoil/agonistas , Factores Trefoil/análisis , Factores Trefoil/antagonistas & inhibidores , Proteínas Supresoras de Tumor/agonistas , Proteínas Supresoras de Tumor/análisis
7.
Cancer Biomark ; 28(2): 141-149, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32116235

RESUMEN

OBJECTIVE: Renal cell carcinoma (RCC) is the most common malignancy involving the kidneys and a major cause of cancer mortality. The involvement of microRNA (miRNA) expression in the tumorigenesis and progression of RCC still not been previously highlighted. We aimed to explore the potential role of miR-222-3p in renal cell carcinoma (RCC). METHOD: We first found that miR-222-3p was elevated significantly in the RCC tissues as compared to the non-tumor counterparts. We also found that a higher level of miR-222-3p in different RCC cell lines than the HK-2 cells. RESULTS: In vitro validation experiment using miR-222-3p mimic molecules significantly induced expression of EMT marker vimentin and downregulated E-cadherin in both 769-P and 786-O RCC cells. In contrary, when miR-222-3p was downregulated by its inhibitor, the reverse observations were made. We then demonstrated a reversal association between the expression level of miR-222-3p and TIMP2/ERK where TIMP2 functions as a tumor suppressor. In a small cohort of 45 clinical samples, we found that miR-222-3p expression level was elevated and was associated with a poorer survival of the patients. Patients with higher miR-222-3p expression showed had a statistically shorter overall survival than those patients of lower miR-222-3p level (HR, 5.789; p= 0.02). CONCLUSION: Collectively, we showed that miR-222-3p functioned as a tumor progression marker and could be a target for future drug development.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Células Renales/genética , Neoplasias Renales/genética , MicroARNs/metabolismo , Inhibidor Tisular de Metaloproteinasa-2/genética , Biomarcadores de Tumor/agonistas , Biomarcadores de Tumor/antagonistas & inhibidores , Carcinoma de Células Renales/mortalidad , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Biología Computacional , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Estimación de Kaplan-Meier , Riñón/patología , Neoplasias Renales/mortalidad , Neoplasias Renales/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Masculino , MicroARNs/agonistas , MicroARNs/antagonistas & inhibidores , Persona de Mediana Edad , Pronóstico
8.
Trends Cancer ; 6(4): 319-336, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32209446

RESUMEN

Circular RNA (circRNA) is a class of single-stranded molecules with tissue/development-specific expression patterns. Unlike linear RNA, circRNA forms a covalently closed loop produced from 'back-splicing' of primary transcripts, conferring on them inherent resistance to exonucleolytic RNA decay. Increasing evidence demonstrates that many circRNAs exert important biological functions by acting as miRNA inhibitors ('sponges'), protein 'decoys', or by encoding small peptides. Importantly, circRNAs are aberrantly expressed in cancer and play indispensable oncogenic or tumor suppressive roles during tumor development and progression. In this review, we summarize the biogenesis, turnover, and involvements of circRNAs in cancer and also discuss their potential as diagnostic biomarkers or therapeutic targets.


Asunto(s)
Antineoplásicos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias/genética , ARN Circular/metabolismo , Animales , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/agonistas , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Progresión de la Enfermedad , Genes Supresores de Tumor , Humanos , Ratones , MicroARNs/metabolismo , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Oncogenes/genética , Precursores del ARN/genética , Empalme del ARN , Estabilidad del ARN , ARN Circular/química , ARN Circular/genética , ARN Interferente Pequeño/farmacología , ARN Interferente Pequeño/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
9.
ACS Nano ; 14(1): 651-663, 2020 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-31851488

RESUMEN

The overexpression of immunomarker programmed cell death protein 1 (PD-1) and engagement of PD-1 to its ligand, PD-L1, are involved in the functional impairment of cluster of differentiation 8+ (CD8+) T cells, contributing to cancer progression. However, heterogeneities in PD-L1 expression and variabilities in biopsy-based assays render current approaches inaccurate in predicting PD-L1 status. Therefore, PD-L1 screening alone is not predictive of patient response to treatment, which motivates us to simultaneously detect multiple immunomarkers engaged in immune modulation. Here, we have developed multimodal probes, immunoactive gold nanostars (IGNs), that accurately detect PD-L1+ tumor cells and CD8+ T cells simultaneously in vivo, surpassing the limitations of current immunoimaging techniques. IGNs integrate the whole-body imaging of positron emission tomography with high sensitivity and multiplexing of Raman spectroscopy, enabling the dynamic tracking of both immunomarkers. IGNs also monitor response to immunotherapies in mice treated with combinatorial PD-L1 and CD137 agonists and distinguish responders from those nonresponsive to treatment. Our results showed a multifunctional nanoscale probe with capabilities that cannot be achieved with either modality alone, allowing multiplexed immunologic tumor profiling critical for predicting early response to immunotherapies.


Asunto(s)
Biomarcadores de Tumor/análisis , Oro/química , Inmunoterapia , Melanoma/diagnóstico por imagen , Melanoma/terapia , Nanopartículas del Metal/química , Imagen Óptica , Animales , Antígeno B7-H1/agonistas , Antígeno B7-H1/análisis , Antígeno B7-H1/genética , Biomarcadores de Tumor/agonistas , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Modelos Animales de Enfermedad , Ratones , Tamaño de la Partícula , Propiedades de Superficie , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/agonistas , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/análisis , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética
10.
Curr Oncol Rep ; 21(11): 97, 2019 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-31696329

RESUMEN

PURPOSE OF REVIEW: In this article, we will briefly review the current treatment landscape for metastatic melanoma and provide a comprehensive update on emerging novel treatment strategies. RECENT FINDINGS: Over the past decade, remarkable advances in immunotherapy and targeted therapy have greatly improved outcomes for patients with advanced melanoma. Although a subset of patients is able to achieve durable responses, the majority experience eventual disease progression on existing therapies. Trials evaluating novel combinatorial strategies, checkpoint inhibitors, immune agonists, T cell-based therapies, intratumoral agents, and others are ongoing. While strides have been made in the treatment of advanced melanoma, further research is needed to identify alternative immune and molecular targets in order to overcome resistance and achieve better clinical outcomes.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Factores Inmunológicos/uso terapéutico , Inmunoterapia , Melanoma/terapia , Terapia Molecular Dirigida , Biomarcadores de Tumor/agonistas , Biomarcadores de Tumor/antagonistas & inhibidores , Resistencia a Antineoplásicos , Humanos , Melanoma/inmunología , Melanoma/patología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
11.
Cancer Biother Radiopharm ; 34(3): 189-195, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30628842

RESUMEN

OBJECTIVE: To investigate the correlations of miR-31 expression with cell proliferation, invasion, and prognosis of patients with head and neck squamous cell carcinoma (HNSCC). METHODS: The expression of miR-31 in human laryngeal cancer TU686 cells, human nasopharyngeal carcinoma CNE-2 cells, and normal human oral keratinocyte (NHOK) epithelial cells was detected via quantitative real-time polymerase chain reaction (qRT-PCR). The effects of miR-31 on the proliferation and invasion of HNSCC cells were explored through transfecting miR-31 analogs (miR-31 mimics) and miR-31 inhibitors (anti-miR-31). qRT-PCR was applied to detect the expressions of miR-31 in 56 cases of HNSCC tumor tissues and tumor-adjacent normal tissues. The correlation of miR-31 expression with pathological parameters and survival prognosis of HNSCC patients was also analyzed. RESULTS: The expressions of miR-31 in TU686 and CNE-2 cell lines were significantly higher than that in NHOK cells (p < 0.01). Compared with those in the negative control group, the proliferation and invasion abilities of cells transfected with miR-31 mimics were notably enhanced (p < 0.01), and those of cells transfected with anti-miR-31 were significantly reduced (p < 0.01). In addition, miR-31 mimics significantly reduced ARID1A expression (p < 0.01) and anti-miR-31 increased its expression (p < 0.05). The expression of miR-31 in tumor tissues of HNSCC patients was remarkably higher than that in tumor-adjacent normal tissues (p < 0.01). This, together with clinical data analysis, revealed that the expression of miR-31 was associated with tumor differentiation, metastasis, and staging of patients, and the survival period of patients with lowly expressed miR-31 was longer. CONCLUSIONS: The highly expressed miR-31 can stimulate the proliferation and invasion of HNSCC cells, closely correlated with tumor differentiation, metastasis, and staging of patients. Patients with lowly expressed miR-31 have a longer survival period. Therefore, miR-31 expression can be taken as a crucial reference indicator for the prognosis of HNSCC patients.


Asunto(s)
Biomarcadores de Tumor/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/genética , MicroARNs/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Apoptosis/efectos de los fármacos , Apoptosis/genética , Biomarcadores de Tumor/agonistas , Biomarcadores de Tumor/antagonistas & inhibidores , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Femenino , Estudios de Seguimiento , Neoplasias de Cabeza y Cuello/mortalidad , Neoplasias de Cabeza y Cuello/patología , Humanos , Masculino , MicroARNs/agonistas , MicroARNs/antagonistas & inhibidores , Persona de Mediana Edad , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Invasividad Neoplásica/prevención & control , Pronóstico , Carcinoma de Células Escamosas de Cabeza y Cuello/mortalidad , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Análisis de Supervivencia
12.
Ann Oncol ; 29(9): 1895-1902, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30137196

RESUMEN

Background: In order to facilitate implementation of precision medicine in clinical management of cancer, there is a need to harmonise and standardise the reporting and interpretation of clinically relevant genomics data. Methods: The European Society for Medical Oncology (ESMO) Translational Research and Precision Medicine Working Group (TR and PM WG) launched a collaborative project to propose a classification system for molecular aberrations based on the evidence available supporting their value as clinical targets. A group of experts from several institutions was assembled to review available evidence, reach a consensus on grading criteria and present a classification system. This was then reviewed, amended and finally approved by the ESMO TR and PM WG and the ESMO leadership. Results: This first version of the ESMO Scale of Clinical Actionability for molecular Targets (ESCAT) defines six levels of clinical evidence for molecular targets according to the implications for patient management: tier I, targets ready for implementation in routine clinical decisions; tier II, investigational targets that likely define a patient population that benefits from a targeted drug but additional data are needed; tier III, clinical benefit previously demonstrated in other tumour types or for similar molecular targets; tier IV, preclinical evidence of actionability; tier V, evidence supporting co-targeting approaches; and tier X, lack of evidence for actionability. Conclusions: The ESCAT defines clinical evidence-based criteria to prioritise genomic alterations as markers to select patients for targeted therapies. This classification system aims to offer a common language for all the relevant stakeholders in cancer medicine and drug development.


Asunto(s)
Biomarcadores de Tumor/genética , Genómica/normas , Oncología Médica/normas , Neoplasias/genética , Medicina de Precisión/métodos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/agonistas , Biomarcadores de Tumor/antagonistas & inhibidores , Biología Computacional/normas , Consenso , Bases de Datos Genéticas/normas , Europa (Continente) , Genómica/métodos , Humanos , Oncología Médica/métodos , Terapia Molecular Dirigida/métodos , Neoplasias/tratamiento farmacológico , Selección de Paciente , Proyectos de Investigación/normas , Sociedades Médicas/normas
13.
Int J Mol Sci ; 19(2)2018 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-29401684

RESUMEN

Acute myeloid leukemias (AML) are clonal disorders of hematopoietic progenitor cells which are characterized by relevant heterogeneity in terms of phenotypic, genotypic, and clinical features. Among the genetic aberrations that control disease development there are microRNAs (miRNAs). miRNAs are small non-coding RNAs that regulate, at post-transcriptional level, translation and stability of mRNAs. It is now established that deregulated miRNA expression is a prominent feature in AML. Functional studies have shown that miRNAs play an important role in AML pathogenesis and miRNA expression signatures are associated with chemotherapy response and clinical outcome. In this review we summarized miRNA signature in AML with different cytogenetic, molecular and clinical characteristics. Moreover, we reviewed the miRNA regulatory network in AML pathogenesis and we discussed the potential use of cellular and circulating miRNAs as biomarkers for diagnosis and prognosis and as therapeutic targets.


Asunto(s)
Biomarcadores de Tumor/genética , Regulación Leucémica de la Expresión Génica , Leucemia Mieloide Aguda/terapia , MicroARNs/genética , Proteínas de Fusión Oncogénica/genética , Animales , Antagomirs/genética , Antagomirs/metabolismo , Antagomirs/uso terapéutico , Biomarcadores de Tumor/agonistas , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/metabolismo , Aberraciones Cromosómicas , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/patología , Humanos , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Ratones , MicroARNs/agonistas , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Terapia Molecular Dirigida , Oligorribonucleótidos/genética , Oligorribonucleótidos/metabolismo , Oligorribonucleótidos/uso terapéutico , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Proteínas de Fusión Oncogénica/metabolismo , Pronóstico , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Cancer Lett ; 402: 203-212, 2017 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-28602976

RESUMEN

Stimulator of interferon genes (STING) is a DNA sensor and an important cytoplasmic adaptor for other DNA sensors, such as Z-DNA binding protein 1 (DAI), DEAD-box helicase 41 (DDX41), and interferon-γ-inducible protein 16 (IFI16). The activation of STING signaling leads to the production of type I interferons and some other pro-inflammatory cytokines, which are critical for host defense against viral infection. Recent accumulating evidences suggest that STING is also involved in tumor development. However, the role of STING signaling in tumorigenesis is complicated, and a comprehensive review is still lacking. In this paper, we provided an overview of the dual role of STING signaling in tumor development from clinical significance to fundamental mechanisms, as well as its pre-clinical application in cancer therapy.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proteínas de la Membrana/metabolismo , Neoplasias/metabolismo , Transducción de Señal , Animales , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/agonistas , Biomarcadores de Tumor/inmunología , Movimiento Celular , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Humanos , Proteínas de la Membrana/agonistas , Proteínas de la Membrana/inmunología , Terapia Molecular Dirigida , Metástasis de la Neoplasia , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neoplasias/patología , Transducción de Señal/efectos de los fármacos
15.
Nutr Cancer ; 68(7): 1202-9, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27552186

RESUMEN

Breast cancer metastasis to the bone continues to be a major health problem, with approximately 80% of advanced breast cancer patients expected to develop bone metastasis. Although the problem of bone metastasis persists, current treatment options for metastatic cancer patients are limited. In this study, we investigated the preventive role of the active vitamin D metabolite, 1α,25-dihydroxyvitamin D (1,25(OH)2D), against the metastatic potential of breast cancer cells using a novel three-dimensional model (rMET) recapitulating multiple steps of the bone metastatic process. Treatment of MCF10CA1a and MDA-MB-231 cells inhibited metastasis in the rMET model by 70% (±5.7%) and 21% (±6%), respectively. In addition, 1,25(OH)2D treatment decreased invasiveness (20 ± 11% of vehicle) and decreased the capability of MCF10CA1a cells to survive in the reconstructed bone environment after successful invasion through the basement membrane (69 ± 5% of vehicle). An essential step in metastasis is epithelial-mesenchymal transition (EMT). Treatment of MCF10CA1a cells with 1,25(OH)2D increased gene (2.04 ± 0.28-fold increase) and protein (1.87 ± 0.20-fold increase) expression of E-cadherin. Additionally, 1,25(OH)2D treatment decreased N-cadherin gene expression (42 ± 8% decrease), a marker for EMT. Collectively, the present study suggests that 1,25(OH)2D inhibits breast cancer cell metastatic capability as well as inhibits EMT, an essential step in the metastatic process.


Asunto(s)
Neoplasias Óseas/metabolismo , Neoplasias de la Mama/metabolismo , Mama/metabolismo , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Vitamina D/análogos & derivados , Anticarcinógenos/metabolismo , Anticarcinógenos/farmacología , Antígenos CD/genética , Antígenos CD/metabolismo , Biomarcadores de Tumor/agonistas , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias Óseas/patología , Neoplasias Óseas/prevención & control , Neoplasias Óseas/secundario , Mama/citología , Mama/efectos de los fármacos , Mama/patología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Cadherinas/agonistas , Cadherinas/antagonistas & inhibidores , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Cinética , Invasividad Neoplásica/patología , Invasividad Neoplásica/prevención & control , Metástasis de la Neoplasia/patología , Metástasis de la Neoplasia/prevención & control , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Vitamina D/metabolismo , Vitamina D/farmacología
16.
Arch Toxicol ; 90(11): 2809-2823, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26542539

RESUMEN

Methyleugenol is a substituted alkenylbenzene found in several herbs and spices. It is classified by the European Union's Scientific Committee on Food as a genotoxic carcinogen. We addressed the biological mechanism of the genotoxic properties of methyleugenol and its oxidative metabolites. Methyleugenol and the oxidative metabolites significantly enhanced the DNA damage in human colon carcinoma cells (HT29). Methyleugenol did not affect the protein status of γH2AX, a biomarker of DNA double-strand breaks, whereas its metabolites methyleugenol-2',3'-epoxide and 3'-oxomethylisoeugenol significantly increased the cellular phosphorylated H2AX level. Both of these metabolites also showed a significant induction of micronuclei in HT29 cells. Furthermore, we investigated whether topoisomerase interaction contribute to the observed effect on DNA integrity. Methyleugenol-2',3'-epoxide and 3'-oxomethylisoeugenol inhibited the activity of recombinant topoisomerase I. In HT29 cells, neither methyleugenol nor the metabolites affected the level of topoisomerase protein bound to DNA, excluding a topoisomerase poisoning mode of action. In addition, 3'-oxomethylisoeugenol potently diminished the level of camptothecin-stabilized topoisomerase I/DNA intermediates and camptothecin-induced DNA strand breaks. In conclusion, it could be suggested that 3'-oxomethylisoeugenol may also interact with classical or food-borne topoisomerase I poisons, diminishing their poisoning effectiveness.


Asunto(s)
Carcinógenos Ambientales/toxicidad , Neoplasias del Colon/inducido químicamente , Daño del ADN , ADN-Topoisomerasas de Tipo I/metabolismo , Eugenol/análogos & derivados , Mutágenos/toxicidad , Inhibidores de Topoisomerasa I/toxicidad , Biomarcadores de Tumor/agonistas , Biomarcadores de Tumor/metabolismo , Biotransformación , Carcinógenos Ambientales/análisis , Carcinógenos Ambientales/metabolismo , Carcinoma/inducido químicamente , Carcinoma/enzimología , Neoplasias del Colon/enzimología , Neoplasias del Colon/metabolismo , Ensayo Cometa , ADN-Topoisomerasas de Tipo I/química , ADN-Topoisomerasas de Tipo I/genética , Compuestos Epoxi/análisis , Compuestos Epoxi/metabolismo , Compuestos Epoxi/toxicidad , Eugenol/análisis , Eugenol/metabolismo , Eugenol/toxicidad , Contaminación de Alimentos , Células HT29 , Histonas/agonistas , Histonas/metabolismo , Humanos , Pruebas de Micronúcleos , Mutágenos/análisis , Mutágenos/metabolismo , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Oxidación-Reducción , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Especias/efectos adversos , Especias/análisis , Inhibidores de Topoisomerasa I/análisis , Inhibidores de Topoisomerasa I/metabolismo
17.
Genome Biol ; 15(8): 453, 2014 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-25222836

RESUMEN

The presence of multiple subclones within tumors mandates understanding of longitudinal and spatial subclonal dynamics. Resolving the spatial and temporal heterogeneity of subclones with cancer driver events may offer insight into therapy response, tumor evolutionary histories and clinical trial design.


Asunto(s)
Biomarcadores de Tumor/agonistas , Biomarcadores de Tumor/genética , Heterogeneidad Genética , Neoplasias/genética , Medicina de Precisión , Evolución Clonal , Humanos
18.
Acta Biomater ; 10(10): 4257-68, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24969665

RESUMEN

Photodynamic therapy (PDT) has attracted much interest for the treatment of cancer due to the increased incidence of multidrug resistance and systemic toxicity in conventional chemotherapy. Phthalocyanine (Pc) is one of main classes of photosensitizers for PDT and possesses optimal photophysical and photochemical properties. A higher specificity can ideally be achieved when Pcs are targeted towards tumor-specific receptors, which may also facilitate specific drug delivery. Herein, we develop a simple and unique strategy to prepare a hydrophilic tumor-targeting photosensitizer ATF-ZnPc by covalently coupling zinc phthalocyanine (ZnPc) to the amino-terminal fragment (ATF) of urokinase-type plasminogen activator (uPA), a fragment responsible for uPA receptor (uPAR, a biomarker overexpressed in cancer cells), through the carboxyl groups of ATF. We demonstrate the high efficacy of this tumor-targeting PDT agent for the inhibition of tumor growth both in vitro and in vivo. Our in vivo optical imaging results using H22 tumor-bearing mice show clearly the selective accumulation of ATF-ZnPc in tumor region, thereby revealing the great potential of ATF-ZnPc for clinical applications such as cancer detection and guidance of tumor resection in addition to photodynamic treatment.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Indoles/farmacología , Neoplasias Experimentales/tratamiento farmacológico , Fotoquimioterapia , Activador de Plasminógeno de Tipo Uroquinasa/farmacología , Animales , Biomarcadores de Tumor/agonistas , Biomarcadores de Tumor/metabolismo , Humanos , Indoles/química , Isoindoles , Ratones , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/agonistas , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Células U937 , Activador de Plasminógeno de Tipo Uroquinasa/química , Ensayos Antitumor por Modelo de Xenoinjerto , Zinc
19.
Cancer Invest ; 30(10): 748-56, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23088770

RESUMEN

IFN regulatory factor 4 binding protein (IBP) has been shown to play an important role in the progression of malignant tumors such as breast cancer cells, but its function in oral squamous cell carcinoma (OSCC) remains unclear. We found that IBP ectopically expressed in some OSCC specimens but not in normal oral mucosa epithelium tissues. IBP expression was significantly correlated with tumor size, differentiation, clinical stage, and distant metastasis. Furthermore, IBP markedly promoted OSCC cell proliferation, shortened the G1 interval in the cell cycle, and increased cyclin D1 expression. These findings suggest that IBP may be a potential therapeutic target for OSCC.


Asunto(s)
Biomarcadores de Tumor/agonistas , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Proteínas de Unión al ADN/biosíntesis , Factores de Intercambio de Guanina Nucleótido/biosíntesis , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Proteínas Nucleares/biosíntesis , Animales , Western Blotting , Línea Celular Tumoral , Proliferación Celular , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos BALB C , Estadificación de Neoplasias , Análisis de Matrices Tisulares , Transfección , Trasplante Heterólogo
20.
J Control Release ; 161(3): 959-66, 2012 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-22617522

RESUMEN

Targeted nanoparticle-based delivery systems have been used extensively to develop effective cancer theranostics. However, how targeting ligands affect extravascular transport of nanoparticles in solid tumors remains unclear. Here, we show, using B16/F10 melanoma cells expressing melanocortin type-1 receptor (MC1R), that the nature of targeting ligands, i.e., whether they are agonists or antagonists, directs tumor uptake and intratumoral distribution after extravasation of nanoparticles from tumor vessels into the extravascular fluid space. Pegylated hollow gold nanospheres (HAuNS, diameter=40 nm) coated with MC1R agonist are internalized upon ligand-receptor binding, whereas MC1R antagonist-conjugated HAuNS remain attached on the cell surface. Transcellular transport of agonist-conjugated HAuNS was confirmed by a multilayer tumor cell model and by transmission electron microscopy. MC1R agonist- but not MC1R antagonist-conjugated nanoparticles exhibit significantly higher tumor uptake than nontargeted HAuNS and are quickly dispersed from tumor vessels via receptor-mediated endocytosis and subsequent transcytosis. These results confirm an active transport mechanism that can be used to overcome one of the major biological barriers for efficient nanoparticle delivery to solid tumors.


Asunto(s)
Oro/administración & dosificación , Melanoma Experimental/metabolismo , Nanosferas/administración & dosificación , Receptor de Melanocortina Tipo 1/metabolismo , Transcitosis/fisiología , Animales , Biomarcadores de Tumor/agonistas , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/metabolismo , Femenino , Oro/química , Células HEK293 , Humanos , Ratones , Ratones Desnudos , Nanosferas/química , Polietilenglicoles/administración & dosificación , Polietilenglicoles/química , Receptor de Melanocortina Tipo 1/agonistas , Receptor de Melanocortina Tipo 1/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...