Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Stem Cells ; 41(8): 762-774, 2023 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-37280108

RESUMEN

Glioblastoma stem cells (GSCs) have unique properties of self-renewal and tumor initiation that make them potential therapeutic targets. Development of effective therapeutic strategies against GSCs requires both specificity of targeting and intracranial penetration through the blood-brain barrier. We have previously demonstrated the use of in vitro and in vivo phage display biopanning strategies to isolate glioblastoma targeting peptides. Here we selected a 7-amino acid peptide, AWEFYFP, which was independently isolated in both the in vitro and in vivo screens and demonstrated that it was able to target GSCs over differentiated glioma cells and non-neoplastic brain cells. When conjugated to Cyanine 5.5 and intravenously injected into mice with intracranially xenografted glioblastoma, the peptide localized to the site of the tumor, demonstrating intracranial tumor targeting specificity. Immunoprecipitation of the peptide with GSC proteins revealed Cadherin 2 as the glioblastoma cell surface receptor targeted by the peptides. Peptide targeting of Cadherin 2 on GSCs was confirmed through ELISA and in vitro binding analysis. Interrogation of glioblastoma databases demonstrated that Cadherin 2 expression correlated with tumor grade and survival. These results confirm that phage display can be used to isolate unique tumor-targeting peptides specific for glioblastoma. Furthermore, analysis of these cell specific peptides can lead to the discovery of cell specific receptor targets that may serve as the focus of future theragnostic tumor-homing modalities for the development of precision strategies for the treatment and diagnosis of glioblastomas.


Asunto(s)
Cadherinas , Técnicas de Visualización de Superficie Celular , Glioblastoma , Péptidos , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Células Madre Neoplásicas , Humanos , Animales , Ratones , Trasplante de Neoplasias , Péptidos/uso terapéutico , Cadherinas/antagonistas & inhibidores , Terapia Molecular Dirigida , Modelos Animales de Enfermedad
2.
Stem Cells ; 39(7): 853-865, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33594762

RESUMEN

Glioblastoma (GBM) ranks among the most lethal of human malignancies with GBM stem cells (GSCs) that contribute to tumor growth and therapeutic resistance. Identification and isolation of GSCs continue to be a challenge, as definitive methods to purify these cells for study or targeting are lacking. Here, we leveraged orthogonal in vitro and in vivo phage display biopanning strategies to isolate a single peptide with GSC-specific binding properties. In silico analysis of this peptide led to the isolation of EYA1 (Eyes Absent 1), a tyrosine phosphatase and transcriptional coactivator. Validating the phage discovery methods, EYA1 was preferentially expressed in GSCs compared to differentiated tumor progeny. MYC is a central mediator of GSC maintenance but has been resistant to direct targeting strategies. Based on correlation and colocalization of EYA1 and MYC, we interrogated a possible interaction, revealing binding of EYA1 to MYC and loss of MYC expression upon targeting EYA1. Supporting a functional role for EYA1, targeting EYA1 expression decreased GSC proliferation, migration, and self-renewal in vitro and tumor growth in vivo. Collectively, our results suggest that phage display can identify novel therapeutic targets in stem-like tumor cells and that an EYA1-MYC axis represents a potential therapeutic paradigm for GBM.


Asunto(s)
Bacteriófagos , Neoplasias Encefálicas , Glioblastoma , Bacteriófagos/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular , Glioblastoma/patología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas/metabolismo
3.
Cell Mol Neurobiol ; 39(8): 1207-1215, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31342232

RESUMEN

The KDEL (Lys-Asp-Glu-Leu) receptors (KDELRs), proteins with seven transmembrane domains, are primarily responsible for endoplasmic reticulum (ER) homeostasis. Recent studies have found additional function of KDELRs in growth, cellular secretory traffic, immune response, and autophagy; however, its role in tumorigenesis is still poorly understood. Here, we showed that KDELR2 is highly expressed in glioblastoma (GBM) tissues. Reviewing the expression of KDELR2 in TCGA and REMBRANDT database, we found that higher expression of KDELR2 is associated with shorter survival of GBM patients. We explored the effect of KDELR2 on tumorigenesis in GBM cells and animal model (nude mice), and identified KDELR2 as oncogene promoting cell proliferation. Additionally, KDELR2 expression in GBM cells correlated positively with HIF1alpha (HIF1α) expression, and we demonstrated by ChIP-qPCR and luciferase reporter assay that the upstream region of the KDELR2 gene is directly targeted by HIF1alpha. Taken together, our data suggest that KDELR2 is a target gene downstream of HIF1-alpha driving the malignancy of GBM and could eventually serve as a therapeutic target for the treatment of GBM patients.


Asunto(s)
Neoplasias Encefálicas/patología , Carcinogénesis/patología , Glioblastoma/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Carcinogénesis/genética , Carcinogénesis/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones Desnudos , Análisis de Supervivencia , Transcripción Genética , Regulación hacia Arriba/genética , Proteínas de Transporte Vesicular/genética
4.
World Neurosurg ; 183: e649-e657, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38181876

RESUMEN

OBJECTIVE: Three-dimensional (3D) printed models are used in the medical field. This study aimed to evaluate the feasibility and safety of a 3D-printed guide plate for use in brain biopsy. METHODS: Twelve patients with intracranial lesions were retrospectively reviewed to determine clinical outcomes and technical procedural operability. These patients underwent brain biopsy assisted with the 3D-printed guide plate. Postoperative computed tomography was performed to assess the accuracy and associated complications of this guide plate. RESULTS: All patients received definite diagnoses assisted by this guide plate. The deviations of the entry and target points were 3.93 ± 0.96 mm and 2.59 ± 0.11 mm, respectively. The angle drift of the puncture path was 5.12° ± 0.14°, and the deviation of the puncture depth was 2.35 ± 1.13 mm. The operation time ranged from 38.5 minutes with local anesthesia to 76.2 minutes with general anesthesia. No patient experienced complications. CONCLUSIONS: The 3D-printed guide plate was noninvasive and had acceptable accuracy and the flexibility of frameless systems. The economic and operative benefits of this device supported its status as a powerful tool for brain biopsy in medical facilities in economically disadvantaged areas or institutions without navigation systems.


Asunto(s)
Encéfalo , Tomografía Computarizada por Rayos X , Humanos , Estudios Retrospectivos , Biopsia/métodos , Encéfalo/diagnóstico por imagen , Encéfalo/cirugía , Encéfalo/patología , Impresión Tridimensional
5.
Front Oncol ; 13: 1124272, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37035164

RESUMEN

Background: Glioblastoma (GBM) is both the most common and aggressive type of primary brain tumor, associated with high mortality rates and resistance to conventional therapy. Despite recent advancements in knowledge and molecular profiling, recurrence of GBM is nearly inevitable. This recurrence has been attributed to the presence of glioma stem cells (GSCs), a small fraction of cells resistant to standard-of-care treatments and capable of self-renewal and tumor initiation. Therefore, targeting these cancer stem cells will allow for the development of more effective therapeutic strategies against GBM. We have previously identified several 7-amino acid length peptides which specifically target GSCs through in vitro and in vivo phage display biopanning. Methods and results: We have combined two of these peptides to create a dual peptide construct (EV), and demonstrated its ability to bind GSCs in vitro and target intracranial GBM in mouse models. A peptide pull-down performed with peptide EV followed by mass spectrometry determined N-cadherin as the binding partner of the peptide, which was validated by enzyme-linked immunosorbent assay and surface plasmon resonance. To develop cytotoxic cellular products aimed at specifically targeting GSCs, chimeric antigen receptors (CARs) were engineered containing the peptide EV in place of the single-chain variable fragment (scFv) as the antigen-binding domain. EV CAR-transduced T cells demonstrated specific reactivity towards GSCs by production of interferon-gamma when exposed to GSCs, in addition to the induction of GSC-specific apoptosis as illustrated by Annexin-V staining. Conclusion: These results exemplify the use of phage display biopanning for the isolation of GSC-targeting peptides, and their potential application in the development of novel cytotoxic therapies for GBM.

6.
J Exp Clin Cancer Res ; 42(1): 238, 2023 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-37697370

RESUMEN

BACKGROUND: Gemcitabine (GEM)-based chemotherapy is the first-line option for pancreatic ductal adenocarcinoma (PDAC). However, the development of drug resistance limits its efficacy, and the specific mechanisms remain largely unknown. RUNX1, a key transcription factor in hematopoiesis, also involved in the malignant progression of PDAC, but was unclear in the chemoresistance of PDAC. METHODS: Comparative analysis was performed to screen GEM-resistance related genes using our single-cell RNA sequencing(scRNA-seq) data and two public RNA-sequencing datasets (GSE223463, GSE183795) for PDAC. The expression of RUNX1 in PDAC tissues was detected by qRT-PCR, immunohistochemistry (IHC) and western blot. The clinical significance of RUNX1 in PDAC was determined by single-or multivariate analysis and survival analysis. We constructed the stably expressing cell lines with shRUNX1 and RUNX1, and successfully established GEM-resistant cell line. The role of RUNX1 in GEM resistance was determined by CCK8 assay, plate colony formation assay and apoptosis analysis in vitro and in vivo. To explore the mechanism, we performed bioinformatic analysis using the scRNA-seq data to screen for the endoplasm reticulum (ER) stress signaling that was indispensable for RUNX1 in GEM resistance. We observed the cell morphology in ER stress by transmission electron microscopy and validated RUNX1 in gemcitabine resistance depended on the BiP/PERK/eIF2α pathway by in vitro and in vivo oncogenic experiments, using ER stress inhibitor(4-PBA) and PERK inhibitor (GSK2606414). The correlation between RUNX1 and BiP expression was assessed using the scRNA-seq data and TCGA dataset, and validated by RT-PCR, immunostaining and western blot. The mechanism of RUNX1 regulation of BiP was confirmed by ChIP-PCR and dual luciferase assay. Finally, the effect of RUNX1 inhibitor on PDAC was conducted in vivo mouse models, including subcutaneous xenograft and patient-derived xenograft (PDX) mouse models. RESULTS: RUNX1 was aberrant high expressed in PDAC and closely associated with GEM resistance. Silencing of RUNX1 could attenuate resistance in GEM-resistant cell line, and its inhibitor Ro5-3335 displayed an enhanced effect in inhibiting tumor growth, combined with GEM treatment, in PDX mouse models and GEM-resistant xenografts. In detail, forced expression of RUNX1 in PDAC cells suppressed apoptosis induced by GEM exposure, which was reversed by the ER stress inhibitor 4-PBA and PERK phosphorylation inhibitor GSK2606414. RUNX1 modulation of ER stress signaling mediated GEM resistance was supported by the analysis of scRNA-seq data. Consistently, silencing of RUNX1 strongly inhibited the GEM-induced activation of BiP and PERK/eIF2α signaling, one of the major pathways involved in ER stress. It was identified that RUNX1 directly bound to the promoter region of BiP, a primary ER stress sensor, and stimulated BiP expression to enhance the reserve capacity for cell adaptation, which in turn facilitated GEM resistance in PDAC cells. CONCLUSIONS: This study identifies RUNX1 as a predictive biomarker for response to GEM-based chemotherapy. RUNX1 inhibition may represent an effective strategy for overcoming GEM resistance in PDAC cells.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Animales , Ratones , Gemcitabina , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Factores de Iniciación de Péptidos , Neoplasias Pancreáticas
7.
Thorac Cancer ; 11(9): 2493-2505, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32686908

RESUMEN

BACKGROUND: Brain metastasis is an unsolved clinical problem in breast cancer patients due to its poor prognosis and high fatality rate. Although accumulating evidence has shown that some pan-histone deacetylase (HDAC) inhibitors can relieve breast cancer brain metastasis, the specific HDAC protein involved in this process is unclear. Thus, identifying a specific HDAC protein closely correlated with breast cancer brain metastasis will not only improve our understanding of the functions of the HDAC family but will also help develop a novel target for precision cancer therapy. METHODS: Immunohistochemical staining of HDAC1, HDAC2, and HDAC3 in 161 samples from breast invasive ductal carcinoma patients, including 63 patients with brain metastasis, was performed using the standard streptavidin-peroxidase method. The relationships between HDAC1, HDAC2, and HDAC3 and overall survival/brain metastasis-free survival/post-brain metastatic survival were evaluated using Kaplan-Meier curves and Cox regression analyses. RESULTS: HDAC1, HDAC2, and cytoplasmic HDAC3 all displayed typical oncogenic characteristics and were independent prognostic factors for the overall survival of breast cancer patients. Only cytoplasmic HDAC3 was an independent prognostic factor for brain metastasis-free survival. Cytoplasmic expression of HDAC3 was further upregulated in the brain metastases compared with the matched primary tumors, while nuclear expression was downregulated. The HDAC1, HDAC2, and HDAC3 expression levels in the brain metastases were not correlated with survival post-brain metastasis. CONCLUSIONS: Our studies first demonstrate a critical role for HDAC3 in the brain metastasis of breast cancer patients and it may serve as a promising therapeutic target for the vigorously developing field of precision medicine. KEY POINTS: Significant findings of the study Cytoplasmic HDAC3 is an independent prognostic factor for the overall survival and brain metastasis-free survival of breast cancer patients. What this study adds Cytoplasmic expression of HDAC3 was further upregulated in the brain metastases compared with the matched primary tumours, while nuclear expression was downregulated.


Asunto(s)
Neoplasias Encefálicas/enzimología , Neoplasias Encefálicas/secundario , Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/enzimología , Histona Desacetilasas/metabolismo , Neoplasias de la Mama/patología , Femenino , Humanos , Persona de Mediana Edad
8.
Int J Cancer ; 125(3): 585-95, 2009 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-19330838

RESUMEN

Malignant gliomas have a tendency to invade diffusely into surrounding healthy brain tissues, thereby precluding their successful surgical removal. The serine/threonine kinase Akt2 is well known as an important regulator of cell survival and growth. In this study, we show that siRNA-mediated depletion of Akt2 inhibited migration and invasion of glioma cells. In addition, we demonstrate the mechanisms by which Akt2 functions to promote cell migration and invasion. Phosphorylation of cofilin, a critical step of actin polymerization, and phosphorylation of Girdin, essential for the integrity of the actin cytoskeleton and cell migration, were impaired. Furthermore, epidermal growth factor-induced ACAP1 phosphorylation and integrin beta1 phosphorylation were also blocked, consistent with defects in adhesion. Thus, Akt2 regulates both cell adhesion and cytoskeleton rearrangement during migration. Decreased MMP-9 expression in Akt2 knocked-down glioma cells was subsequently confirmed by Western blotting, consistent with the decreased invasion in vitro and in vivo. These results suggest that Akt2 contributes to glioma cells migration and invasion by regulating the formation of cytoskeleton, influencing adhesion and increasing expression of MMP-9. Our immunohistochemistry results by using human gliomas tissue sections also indicated that Akt2 expression was closely related with the malignancy of gliomas. This is coincident with our in vivo and in vitro results from cell lines. All of these results indicate that Akt2 is a critical factor in gliomas invasion. This study identifies that Akt2 is a potentially antiinvasion target for therapeutic intervention in gliomas.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Movimiento Celular , Glioma/metabolismo , Glioma/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Actinas/metabolismo , Animales , Western Blotting , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular , Quimiotaxis , Proteínas Activadoras de GTPasa/metabolismo , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Inmunohistoquímica , Inmunoprecipitación , Integrina beta1/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Proteínas de Microfilamentos/metabolismo , Microscopía Fluorescente , Invasividad Neoplásica , Fosforilación , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Transfección , Proteínas de Transporte Vesicular/metabolismo
9.
Thorac Cancer ; 10(2): 137-142, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30485664

RESUMEN

BACKGROUND: Brain metastases (BM) are a common consequence of lung cancer and surgery is effective; however, the factors affecting survival after surgery are unclear. The aim of this study was to identify the outcomes and prognoses of post-metastasectomy patients with BM from non-small cell lung cancer (NSCLC) at a single institution over a 15-year period. METHODS: NSCLC patients who had undergone BM surgery were retrospectively identified. Survival was analyzed using the Kaplan-Meier curve, and univariate and multivariate factors associated with survival were identified using the Cox proportional hazards model. RESULTS: The median overall survival was 9.8 months, 18 (14.8%) patients survived > 24 months, and 6 (4.9%) > 36 months. The one and two-year survival rates were 41% and 18.6%, respectively. Univariate analysis revealed that recursive partitioning analysis (RPA) classification, Karnofsky Performance Scale (KPS) scores, BM number, extracranial metastasis status, different lesion locations, resection extent, postoperative treatment, and salvage therapy after recurrence significantly influenced patient survival. The different treatment modalities for primary lesions also affected postoperative survival. KPS ≥ 70, RPA class I/II, and postoperative chemotherapy were independent factors that decreased the risk of death from BM. Interestingly, the initial onset of intracranial lesions could increase the risk of death from BM. CONCLUSION: A KPS score ≥ 70, RPA class I/II, and postoperative chemotherapy could benefit post-metastasectomy patients with BM from NSCLC. Conversely, the initial onset of intracranial lesions is an unfavorable factor that increases the risk of death. These findings support the use of personalized therapy for patients with BM from NSCLC.


Asunto(s)
Adenocarcinoma del Pulmón/mortalidad , Neoplasias Encefálicas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Células Escamosas/mortalidad , Neoplasias Pulmonares/mortalidad , Neumonectomía/mortalidad , Adenocarcinoma del Pulmón/patología , Adenocarcinoma del Pulmón/cirugía , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Encefálicas/secundario , Neoplasias Encefálicas/cirugía , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/cirugía , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/cirugía , Femenino , Estudios de Seguimiento , Humanos , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/cirugía , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Tasa de Supervivencia
10.
Cancer Biol Med ; 16(3): 606-617, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31565489

RESUMEN

OBJECTIVE: More than half of human glioblastomas show EGFR gene amplification and mutation, but EGFR inhibitors have not been effective in treating EGFR-positive glioblastoma patients. The mechanism behind this type of primary resistance is not well understood. The aim of this study was to investigate gefitinib resistance in glioblastoma, and explore ways to circumvent this significant clinical problem. METHODS: MTT method was used to test the cell viability after EGFR-positive glioblastoma cells were treated with indicated drugs; real-time quantitative PCR method was included to detect the TNFα mRNA levels in glioma tissues and cell lines. ELISA was introduced to measure the TNFα protein levels in cell culture supernatant of glioblastoma cells treated with gefitinib. Western blot was used to detect the activity change of intracellular kinases in drug-treated glioblastoma cells. Two mouse xenograft tumor models were carried out to evaluate the in vivo effects of a combination of EGFR and TNFα inhibitors. RESULTS: We found that glioblastoma resistance to gefitinib may be mediated by an adaptive pro-survival TNFα-JNK-Axl signaling axis, and that high TNFα levels in the glioblastoma microenvironment may further intensify primary resistance. A combination of the TNFα-specific small-molecule inhibitor C87 and gefitinib significantly enhanced the sensitivity of glioblastoma cells to gefitinib in vitro and in vivo. CONCLUSIONS: Our findings provide a possible explanation for the primary resistance of glioblastoma to EGFR inhibitors and suggest that dual blockade of TNFα and EGFR may be a viable therapeutic strategy for the treatment of patients with chemotherapy-refractory advanced glioblastoma.

11.
Cancer Biol Med ; 14(2): 183-186, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28607809

RESUMEN

Non-small cell lung cancer (NSCLC) ranks as the leading cause of cancer-related death in the world. Brain metastasis (BM) is a common complication of NSCLC, with 25%-40% of patients developing BM during the course of the disease. A significant strategy of local disease control in the central nervous system is radiation therapy. With the development of precision medicine, the concept of treating lung cancer BM has gradually changed. In this case, we performed a surgical procedure to obtain enough tumor tissue for the detection of the target gene and other related experiments after the patient was informed. Finally, we found that the patient had both hepatocyte growth factor receptor (MET) gene amplification and kinesin light chain 1-anaplastic lymphoma kinase fusion (KLC1-ALK) through next-generation sequencing and showed sensitivity to the targeted therapy of crizotinib. The patient exhibited good response. Our case was successful and underwent targeted therapy with the guidance of precise diagnosis.

12.
Oncol Lett ; 11(4): 2353-2358, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27073479

RESUMEN

Previous in vivo and in vitro studies have shown that human mesenchymal stem cells (MSCs) exhibit tropism for gliomas. However, the mechanism underlying this directed migration remains unclear. The aim of the present study was to investigate the possible mechanism underlying platelet-derived growth factor-BB (PDGF-BB)-induced chemotactic migration of bone marrow-derived MSCs (BMSCs) toward glioma. Rat glioma C6 cell-conditioned medium was utilized to evaluate the chemotactic response of BMSCs toward glioma using an in vitro migration assay. Recombinant rat PDGF-BB was added to C6 cell-conditioned medium to assess its effect on the tropism of BMSCs. The effect of PDGF-BB on the expression levels of cluster of differentiation (CD)44 in BMSCs was evaluated by reverse transcription-polymerase chain reaction (RT-PCR) and immunofluorescence assays. The results revealed that chemotactic migration was induced in BMSCs by rat glioma C6 cell-conditioned medium, which was enhanced by PDGF-BB treatment in a dose-dependent manner. Furthermore, RT-PCR and immunofluorescence assays showed that CD44 expression was upregulated in BMSCs following treatment with 40 ng/ml PDGF-BB for 12 h. Additionally, 3-h pretreatment with the anti-CD44 neutralizing antibody OX-50 was observed to attenuate the tropism of BMSCs toward glioma in the presence or absence of PDGF-BB. The results of the present study indicate that CD44 mediates the tropism of BMSCs toward glioma, and PDGF-BB promotes the migration of BMSCs toward glioma via the upregulation of CD44 expression in BMSCs. These findings suggest CD44 inhibition may be a potential therapeutic target for the treatment of glioma.

13.
Zhonghua Xue Ye Xue Za Zhi ; 36(4): 282-5, 2015 Apr.
Artículo en Zh | MEDLINE | ID: mdl-25916286

RESUMEN

OBJECTIVE: To explore clinical characteristics, treatment and prognosis of primary central nervous system lymphoma(PCNSL). METHODS: Retrospective analysis, Kaplan-Meier analysis and Log-rank test were conducted on 30 PCNSL patients from 2006 to 2014 in our hospital. RESULTS: The median age of this cohort (14 males and 16 females) was 57.4 years old. 18 cases had single tumor, 12 cases multiple. 17 cases presentd with intracranial hypertension and 13 cases focal neurological deficits. 13 cases (62%) were diffuse large B cell lymphoma. About 60% patients received combination therapy including surgery, radiotherapy or chemotherapy. 63.3% complete remission rate (CR) was achieved for all patients. Kaplan-Meier analysis and Log-rank test showed the median overall survival (OS) was 24 months, the rates of 2-year survival, 5-year survival, 6-month progression-free survival(PFS) and 1-year free-progressed survival (PFS) were as of 46.7%, 13.3%, 60.0% and 43.3% respectively. The median OS of 11 patients received whole brain radiotherapy(WBRT)combined with chemotherapy was 48 months. The median OS of 7 patients treated with stereotactic radiosurgery(SRS) combined with chemotherapy had no significant difference when compared to the former (P=0.233). Survive analysis showed that age was prognostic factor for PCNSL patients(P=0.030). CONCLUSION: Diffuse large B cell lymphoma was the main type of PCNSL, single or multiple location, presented with increased intracranial hypertension or focal neurological deficits. Age was the key prognostic factor for patients. Surgery was suitable for patients with supertentorial and superficial tumor or with acute intracranial hypertension syndrome. SRS was a feasible local therapy which alleviated the symptoms and led to less toxicity. PCNSL patients might benefit from multimode therapy.


Asunto(s)
Neoplasias del Sistema Nervioso Central , Linfoma , Terapia Combinada , Supervivencia sin Enfermedad , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Pronóstico , Inducción de Remisión , Estudios Retrospectivos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA