Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 128
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Lab Invest ; 99(10): 1484-1500, 2019 10.
Article in English | MEDLINE | ID: mdl-31201367

ABSTRACT

A previous study revealed that therapeutic miR-26a delivery suppresses tumorigenesis in a murine liver cancer model, whereas we found that forced miR-26a expression increased hepatocellular carcinoma (HCC) cell migration and invasion, which prompted us to characterize the causes and mechanisms underlying enhanced invasion due to ectopic miR-26a expression. Gain-of-function and loss-of-function experiments demonstrated that miR-26a promoted migration and invasion of BEL-7402 and HepG2 cells in vitro and positively modulated matrix metalloproteinase (MMP)-1, MMP-2, MMP-9, and MMP-10 expression. In addition, exogenous miR-26a expression significantly enhanced the metastatic ability of HepG2 cells in vivo. miR-26a negatively regulated in vitro proliferation of HCC cells, and miR-26a overexpression suppressed HepG2 cell tumor growth in nude mice. Further studies revealed that miR-26a inhibited cell growth by repressing the methyltransferase EZH2 and promoted cell migration and invasion by inhibiting the phosphatase PTEN. Furthermore, PTEN expression negatively correlated with miR-26a expression in HCC specimens from patients with and without metastasis. Thus, our findings suggest for the first time that miR-26a promotes invasion/metastasis by inhibiting PTEN and inhibits cell proliferation by repressing EZH2 in HCC. More importantly, our data also suggest caution if miR-26a is used as a target for cancer therapy in the future.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Liver Neoplasms/metabolism , MicroRNAs/metabolism , PTEN Phosphohydrolase/metabolism , Animals , Cell Movement , Female , Hep G2 Cells , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis
2.
Biochem Biophys Res Commun ; 482(2): 253-256, 2017 Jan 08.
Article in English | MEDLINE | ID: mdl-27847318

ABSTRACT

Nuclear receptor coactivator 5 (NCOA5) is known to modulate ERα-mediated transcription and has been found to be involved in the progression of several malignancies. However, the potential correlation between NCOA5 and clinical outcome in patients with luminal breast cancer remains unknown. In the present study, we demonstrated that NCOA5 was significantly up-regulated in luminal breast cancer tissues compared with adjacent non-cancerous tissues both in validated cohort and TCGA cohort. Moreover, Kaplan-Meier analysis indicated that patients with high NOCA5 expression had significantly lower overall survival (P = 0.021). Cox regression analysis indicated that the high NOCA5 expression was independent high risk factor as well as old age (>60) and HER-2 expression (P = 0.039; P = 0.003; P = 0.005; respectively). This study provides new insights and evidences that NOCA5 over-expression was significantly correlated with progression and prognosis in luminal breast cancer. However, the precise cellular mechanisms for NOCA5 in luminal breast cancer need to be further explored.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Nuclear Receptor Coactivators/metabolism , Age Distribution , Aged , Aged, 80 and over , Breast Neoplasms/diagnosis , China/epidemiology , Disease Progression , Female , Humans , Incidence , Middle Aged , Prognosis , Reproducibility of Results , Risk Factors , Sensitivity and Specificity , Statistics as Topic , Survival Rate
3.
Biochem Biophys Res Commun ; 477(4): 923-926, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27378428

ABSTRACT

Myeloid leukemia factor 1-interacting protein (MLF1IP) has been found to be involved in the progression of several malignancies. The potential correlation between MLF1IP and clinical outcome in patients with luminal breast cancer, however, remains unknown. In the present study, we demonstrated that MLF1IP was significantly upregulated in luminal breast cancer tissue compared with adjacent normal tissue both in validated cohort and TCGA cohort. Upregulated expression of MLF1IP was correlated with more often lymph node metastasis and negative progesterone receptor expression in TCGA cohorts. Kaplan-Meier analysis indicated that patients with high MLF1IP expression had significantly lower overall survival. Moreover, multivariate analysis revealed that high MLF1IP expression was independent high risk factor as well as old age (>60) and distant metastasis. This study provides new insights and evidences that MLF1IP over-expression plays important roles in progression of luminal breast cancer. However, the precise cellular mechanisms for MLF1IP in luminal breast cancer need to be further explored.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Nuclear Proteins/metabolism , Age Distribution , Aged , Aged, 80 and over , Breast Neoplasms/pathology , Cell Cycle Proteins , China/epidemiology , Disease Progression , Female , Histones , Humans , Middle Aged , Neoplasm Invasiveness , Prevalence , Prognosis , Reproducibility of Results , Risk Assessment , Sensitivity and Specificity , Survival Rate
4.
Ann Surg Oncol ; 22(1): 32-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25012266

ABSTRACT

PURPOSE: This study was designed to examine the relationship between different methodologies for response evaluation and long-term survival estimation in patients underwent neoadjuvant chemotherapy (NCT) for breast cancer. METHODS: We retrospectively analyzed 569 patients who were diagnosed with LABC and received NCT followed by breast and axilla surgery. The RECIST 1.1 criteria and Miller-Payne (MP) grading scale were used to evaluate patient responses to NCT. Univariate and multivariate survival analyses were performed to investigate the correlation between treatment response and long-term patient survival. RESULTS: Clinical response (RFS [P < 0.001]; OS [P = 0.003]), pathological response evaluated by pCR (RFS [P < 0.001]; OS [P < 0.001]), and MP grade (RFS [P < 0.001]; OS [P < 0.001]) were significant predictors of risks of relapse and survival. However, in hormone receptor-positive (ER and/or PR+) subtypes, the clinical response (P = 0.004 for Luminal-A and P = 0.038 for Luminal-B) and MP grade (P = 0.002 for Luminal-A and P < 0.001 for Luminal-B) significantly predicted RFS independently according to multivariate Cox regression model. MP grade (P = 0.015 for Luminal-A and P = 0.009 for Luminal-B) also was an independent predictor of patients' OS. However, these two methods failed to predict patient survival in hormone receptor-negative (ER and PR-) subtypes. CONCLUSIONS: Our findings indicate that the value of response evaluation methods varies for different breast cancer subtypes. Conceiving of further prospective approaches for new individualized response-evaluation models are needed in the neoadjuvant setting.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Neoadjuvant Therapy/mortality , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology , Adult , Aged , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Chemotherapy, Adjuvant , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , Middle Aged , Neoplasm Grading , Neoplasm Staging , Prognosis , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Retrospective Studies , Survival Rate , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Young Adult
5.
BMC Genet ; 15: 67, 2014 Jun 11.
Article in English | MEDLINE | ID: mdl-24919593

ABSTRACT

BACKGROUND: Aberrant hypermethylation of gene promoter regions is a primary mechanism by which tumor suppressor genes become inactivated in breast cancer. Epigenetic inactivation of the protein tyrosine phosphatase receptor-type O gene (PTPRO) has been described in several types of cancer. RESULTS: We screened primary breast cancer tissues for PTPRO promoter hypermethylation and assessed potential associations with pathological features and patient outcome. We also evaluated its potential as a breast cancer biomarker. PTPRO methylation was observed in 53 of 98 (54%) breast cancer tissues but not in adjacent normal tissue. Among matched peripheral blood samples from breast cancer patients, 33 of 98 (34%) exhibited methylated PTPRO in plasma. In contrast, no methylated PTPRO was observed in normal peripheral blood from 30 healthy individuals. PTPRO methylation was positively associated with lymph node involvement (P = 0.014), poorly differentiated histology (P = 0.037), depth of invasion (P = 0.004), and HER2 amplification (P = 0.001). Multivariate analysis indicated that aberrant PTPRO methylation could serve as an independent predictor for overall survival hazard ratio (HR): 2.7; 95% CI: 1.1-6.2; P = 0.023), especially for patients with HER2-positive (hazard ratio (HR): 7.5; 95% CI: 1.8-31.3; P = 0.006), but not in ER + and PR + subpopulation. In addition, demethylation induced by 5-azacytidine led to gene reactivation in PTPRO-methylated and -silenced breast cancer cell lines. CONCLUSIONS: Here, we report that tumor PTPRO methylation is a strong prognostic factor in breast cancer. Methylation of PTPRO silences its expression and plays an important role in breast carcinogenesis. The data we present here may provide insight into the development of novel therapies for breast cancer treatment. Additionally, detection of PTPRO methylation in peripheral blood of breast cancer patients may provide a noninvasive means to diagnose and monitor the disease.


Subject(s)
Biomarkers, Tumor/blood , Breast Neoplasms/genetics , DNA Methylation , Promoter Regions, Genetic , Receptor-Like Protein Tyrosine Phosphatases, Class 3/blood , Breast Neoplasms/diagnosis , Cell Line, Tumor , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , Humans , Prognosis
6.
Tumour Biol ; 34(1): 555-63, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23188703

ABSTRACT

X-ray repair cross-complementing group 1 (XRCC1), a DNA repair enzyme, plays a crucial role in the base excision repair by generating a single nucleotide repair patch. It has been demonstrated that the XRCC1 Arg399Gln gene polymorphism was associated with variations in XRCC1 enzyme activity. The aim of this study was to quantitatively summarize the association between the XRCC1 Arg399Gln polymorphism and susceptibility to colorectal cancer (CRC). A comprehensive search of the PubMed, Embase, and China National Knowledge Infrastructure databases was conducted for studies on the association between the XRCC1 Arg399Gln polymorphism and CRC risk. Summary odds ratio (OR) with its corresponding 95 % confidence interval (95 %CI) was estimated, in a fixed-effects model or a random-effects model when appropriate, to assess the association. Totally, 26 case-control studies with 6,979 cases and 11,470 controls were included into this meta-analysis. The pooled results of total studies showed that the XRCC1 Arg399Gln polymorphism was significantly associated with increased risk of CRC in all genetic contrast models (OR(A vs. G) = 1.13, 95 %CI 1.03-1.23, P (OR) = 0.008; OR(Gln/Gln vs. Arg/Arg) = 1.24, 95 %CI 1.04-1.46, P (OR) = 0.015; OR(Gln/Gln vs. Arg/Gln + Arg/Arg) = 1.19, 95 %CI 1.03-1.38, P (OR) = 0.021; OR(Gln/Gln + Arg/Gln vs. Arg/Arg) = 1.14, 95 %CI 1.02-1.28, P (OR) = 0.022), except for the additive contrast model (OR(Arg/Gln vs. Arg/Arg) = 1.11, 95 %CI 0.99-1.25, P (OR) = 0.064). The statistically significant association between the XRCC1 Arg399Gln polymorphism and CRC risk was observed among studies with high quality and in Asians, but not in Caucasians. Sensitivity analyses by sequential omission of any individual studies further identified the significant association. Publication bias was inexistent in this meta-analysis. The meta-analysis suggests that the XRCC1 Arg399Gln polymorphism is associated with increased risk of CRC.


Subject(s)
Colorectal Neoplasms/genetics , DNA-Binding Proteins/genetics , Genetic Predisposition to Disease/ethnology , Asian People , Case-Control Studies , Colorectal Neoplasms/ethnology , DNA Repair , Humans , Polymorphism, Single Nucleotide , White People , X-ray Repair Cross Complementing Protein 1
7.
Mol Biol Rep ; 39(2): 1113-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21603857

ABSTRACT

Epidemiological studies have evaluated the association between ATM 5557G>A (p.D1853N) polymorphism and breast cancer risk. However, the results remain conflicting rather than conclusive. To derive a more precise estimation of the relationship, we performed this meta-analysis. Systematic searches of PubMed and Medline databases were performed. A total of nine studies included 3155 cases and 2752 controls were identified. When all nine studies were pooled into the meta-analysis, there was no evidence for significant association between 5557G>A mutation and breast cancer risk(for G/A vs. G/G: OR = 1.05, 95% CI = 0.83-1.34; for A/A vs. G/G: OR = 0.77, 95% CI = 0.58-1.03; for dominant model: OR = 1.04, 95% CI = 0.82-1.31; for recessive model: OR = 0.87, 95% CI = 0.69-1.09). In the subgroup analyses by family history and ethnicity, significant associations were found among Amerindians (for G/A vs. G/G: OR = 2.19, 95% CI = 1.38-3.47; for dominant model: OR = 2.15, 95% CI = 1.37-3.38). In summary, the meta-analysis suggest that ATM 5557G>A polymorphism is associated with increased breast cancer risk among Amerindians. However, due to the small subjects included in analysis and the selection bias existed in some studies, the results for Amerindians should be interpreted with caution.


Subject(s)
Breast Neoplasms/epidemiology , Breast Neoplasms/genetics , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Indians, South American/genetics , Polymorphism, Single Nucleotide/genetics , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/genetics , Ataxia Telangiectasia Mutated Proteins , Female , Genetic Association Studies , Humans , Linear Models , Odds Ratio , Risk Factors
8.
Cochrane Database Syst Rev ; (7): CD008926, 2012 Jul 11.
Article in English | MEDLINE | ID: mdl-22786516

ABSTRACT

BACKGROUND: Toremifene (TOR) and tamoxifen (TAM) can both be used as treatments for advanced breast cancer. OBJECTIVES: To compare the efficacy and safety of TOR with TAM in patients with advanced breast cancer. SEARCH METHODS: The Cochrane Breast Cancer Group's Specialised Register was searched (1 July 2011) using the codes for "toremifene", "fareston", "tamoxifen, "nolvadex, and "breast cancer". We also searched MEDLINE (via PubMed) (from inception to 1 July 2011), EMBASE (via Ovid) (from inception to 1 July 2011), The Cochrane Central Register of Controlled Trials (CENTRAL) (The Cochrane Library, Issue 7, 2011), and the WHO International Clinical Trials Registry Platform search portal (1 July 2011). In addition, we screened the reference lists of relevant trials or reviews. SELECTION CRITERIA: Randomised controlled trials (RCTs) that compared the efficacy and safety, or both of TOR with TAM in women with advanced breast cancer. Trials that provided sufficient data on one of the following items: objective response rate (ORR), time to progression (TTP), overall survival (OS), and adverse events, were considered eligible for inclusion. DATA COLLECTION AND ANALYSIS: Studies were assessed for eligibility and quality. Two review authors independently extracted the following details: first author, publication year, country, years of follow-up, treatment arms, intention-to-treat (ITT) population size, menopausal status of patients, hormone receptor status, response criteria, efficacy and safety outcomes of TOR and TAM arms. Hazard ratios (HR) were derived for time-to-event outcomes, where possible, and response and adverse events were analysed as dichotomous variables. We used a fixed-effect model for meta-analysis unless there was significant between-study heterogeneity. MAIN RESULTS: A total of 2061 patients from seven RCTs were included for final analysis, with 1226 patients in the TOR group and 835 patients in the TAM group. The ORR for the TOR group was 25.8% (316/1226) whereas, the ORR for the TAM group was 26.9% (225/835). The pooled risk ratio (RR) suggested that the ORRs were not statistically different between the two groups (RR 1.02, 95% confidence interval (CI) 0.88 to 1.18, P = 0.83). The median TTP was 6.1 months for the TOR group and 5.8 months for the TAM group. The median OS was 27.8 months for the TOR group and 27.6 months for the TAM group. There were no significant differences in TTP and OS between the two therapeutic groups (for TTP: HR 1.08, 95% CI 0.94 to 1.24; for OS: HR 1.02, 95% CI 0.86 to 1.20). The frequencies of most adverse events were also similar in the two groups, while headache seemed to occur less in the TOR group than in the TAM group (RR 0.14, 95% CI 0.03 to 0.74, P = 0.02). There was no significant heterogeneity between studies in most of the above meta-analyses. Sensitivity analysis did not alter the results. AUTHORS' CONCLUSIONS: TOR and TAM are equally effective and the safety profile of the former is at least not worse than the latter in the first-line treatment of patients with advanced breast cancer. Thus, TOR may serve as a reasonable alternative to TAM when anti-oestrogens are applicable but TAM is not the preferred choice for some reason.


Subject(s)
Antineoplastic Agents, Hormonal/adverse effects , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Tamoxifen/therapeutic use , Toremifene/therapeutic use , Aged , Female , Humans , Middle Aged , Randomized Controlled Trials as Topic , Tamoxifen/adverse effects , Toremifene/adverse effects
9.
Zhonghua Zhong Liu Za Zhi ; 33(8): 574-8, 2011 Aug.
Article in Zh | MEDLINE | ID: mdl-22325215

ABSTRACT

OBJECTIVE: To explore the changes of the sensitivity of cetuximab-resistant human nasopharyngeal carcinoma (hNPC) cells 5-8F/Erbitux to cetuximab by silencing H-ras gene with RNA interference (RNAi). METHODS: The 5-8F/Erbitux cells were induced by stepwise exposure to increasing doses of cetuximab. Western blot was conducted to detect the protein levels of H-ras and K-ras. Real-time PCR was employed to detect the expression of H-ras and K-ras. H-ras-shRNA plasmids (shRNA vector carrying the H-ras gene) were constructed and transferred into 5-8F/Erbitux cells. The gene and protein expression levels of H-ras and the changes of the sensitivity of 5-8F/Erbitux cells to cetuximab after transfection were measured, respectively. RESULTS: After treatment with cetuximab for 3 and 5 days, the resistance index (RI) of the 5-8F/Erbitux cells was 1.2 and 1.1, and the protein levels of H-ras and K-ras in 5-8F/Erbitux cells were 0.798 +/- 0.019 and 0.190 +/- 0.011, respectively, significantly higher than that in the 5-8F cells (P<0.001). The gene expressions of H-ras and K-ras in 5-8F/Erbitux cells were 1.260 +/- 0.114 and 0.850 +/- 0.006, respectively. Compared with 5-8F cells, the former was higher (P = 0.016) and the latter was lower (P = 0.000). After transfection with H-ras-shRNA plasmid, the 5-8F/Erbitux cells showed reduced levels of H-ras gene and protein, and the cell apoptosis and inhibition rates increased significantly (P<0.001). CONCLUSIONS: H-ras siRNA can reverse cetuximab-resistance of 5-8F/Erbitux cells through down-regulation of H-ras gene expression, indicating that the generation of cetuximab-resistance in 5-8F/Erbitux cells is associated with amplification and overexpression of the H-ras gene.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Nasopharyngeal Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , RNA Interference , Antibodies, Monoclonal, Humanized , Apoptosis , Carcinoma , Cell Line, Tumor , Cell Proliferation , Cetuximab , Gene Expression Regulation, Neoplastic , Genes, ras , Humans , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Plasmids , RNA, Small Interfering/genetics , Transfection
10.
Chin J Cancer ; 30(10): 682-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21959045

ABSTRACT

The efficacy and safety of bevacizumab with modified irinotecan, leucovorin bolus, and 5-fluorouracil intravenous infusion (mIFL) in the first-line treatment of metastatic colorectal cancer (mCRC) has not been well evaluated in randomized clinical trials in Chinese patients. We conducted a phrase III trial in which patients with previously untreated mCRC were randomized 2:1 to the mIFL [irinotecan (125 mg/m(2)), leucovorin (20 mg/m(2)) bolus, and 5-fluorouracil intravenous infusion (500 mg/m(2)) weekly for four weeks every six weeks] plus bevacizumab (5 mg/kg every two weeks) group and the mIFL group, respectively. Co-primary objectives were progression-free survival (PFS) and 6-month PFS rate. In total, 214 patients were enrolled. Our results showed that addition of bevacizumab to mIFL significantly improved median PFS (4.2 months in the mIFL group vs. 8.3 months in the bevacizumab plus mIFL group, P < 0.001), 6-month PFS rate (25.0% vs. 62.6%, P < 0.001), median overall survival (13.4 months vs. 18.7 months, P = 0.014), and response rate (17% vs. 35%, P = 0.013). Grades 3 and 4 adverse events included diarrhea (21% in the mIFL group and 26% in the bevacizumab plus mIFL group) and neutropenia (19% in the mIFL group and 33% in the bevacizumab plus mIFL group). No wound-healing complications or congestive heart failure occurred. Our results suggested that bevacizumab plus mIFL is effective and well tolerated as first-line treatment for Chinese patients with mCRC. Clinical benefit and safety profiles were consistent with those observed in pivotal phase III trials with mainly Caucasian patients.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Adult , Aged , Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Asian People , Bevacizumab , Camptothecin/administration & dosage , Camptothecin/adverse effects , Camptothecin/analogs & derivatives , Colorectal Neoplasms/pathology , Diarrhea/chemically induced , Disease-Free Survival , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Humans , Irinotecan , Leucovorin/administration & dosage , Leucovorin/adverse effects , Male , Middle Aged , Neoplasm Metastasis , Neutropenia/chemically induced , Prospective Studies , Survival Rate , Young Adult
11.
Chin J Cancer ; 30(5): 327-35, 2011 May.
Article in English | MEDLINE | ID: mdl-21527065

ABSTRACT

Overexpression of human epidermal growth factor receptor-2 (HER2) in metastatic breast cancer (MBC) is associated with poor prognosis. This single-arm open-label trial (EGF109491; NCT00508274) was designed to confirm the efficacy and safety of lapatinib in combination with capecitabine in 52 heavily pretreated Chinese patients with HER2-positive MBC. The primary endpoint was clinical benefit rate (CBR). Secondary endpoints included progression-free survival (PFS), time to response (TTR), duration of response (DoR), central nervous system (CNS) as first site of relapse, and safety. The results showed that there were 23 patients with partial responses and 7 patients with stable disease, resulting in a CBR of 57.7%. The median PFS was 6.34 months (95% confidence interval, 4.93-9.82 months). The median TTR and DoR were 4.07 months (range, 0.03-14.78 months) and 6.93 months (range, 1.45-9.72 months), respectively. Thirteen (25.0%) patients had new lesions as disease progression. Among them, 2 (3.8%) patients had CNS disease reported as the first relapse. The most common toxicities were palmar-plantar erythrodysesthesia (59.6%), diarrhea (48.1%), rash (48.1%), hyperbilirubinemia (34.6%), and fatigue (30.8%). Exploratory analyses of oncogenic mutations of PIK3CA suggested that of 38 patients providing a tumor sample, baseline PIK3CA mutation status was not associated with CBR (P = 0.639) or PFS (P = 0.989). These data confirm that the lapatinib plus capecitabine combination is an effective and well-tolerated treatment option for Chinese women with heavily pretreated MBC, irrespective of PIK3CA status.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Deoxycytidine/analogs & derivatives , Fluorouracil/analogs & derivatives , Quinazolines/administration & dosage , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Asian People , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Capecitabine , Class I Phosphatidylinositol 3-Kinases , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Diarrhea/chemically induced , Disease Progression , Disease-Free Survival , Exanthema/chemically induced , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Hand-Foot Syndrome/etiology , Humans , Lapatinib , Middle Aged , Mutation , Neoplasm Staging , Phosphatidylinositol 3-Kinases/genetics , Quinazolines/adverse effects , Receptor, ErbB-2/metabolism , Remission Induction
12.
Front Public Health ; 9: 671788, 2021.
Article in English | MEDLINE | ID: mdl-34109148

ABSTRACT

Coronavirus disease 2019 (COVID-19) is caused by a new coronavirus, namely severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and is currently spreading all over the world. In this paper, we developed a practical model for identifying the features of cytokine storm, which is common in acute infectious diseases and harmful manifestation of COVID-19, by distinguishing major and minor clinical events. This model is particularly suitable for identifying febrile and infectious diseases like COVID-19. Based on this model, features of cytokine storm and pathogenesis of COVID-19 have been proposed to be a consequence of the disequilibrated cytokine network resulting from increased biological activity of transforming growth factor-ß (TGF-ß), which induces certain clinical manifestations such as fatigue, fever, dry cough, pneumonia, abatement and losing of olfactory, and taste senses in some patients. Research and clarification of the pathogenesis of COVID-19 will contribute to precision treatment. Various anti-TGF-ß therapies may be explored as potential COVID-19 treatment. This novel model will be helpful in reducing the widespread mortality of COVID-19.


Subject(s)
COVID-19 Drug Treatment , Coronavirus Infections , Cytokine Release Syndrome , Humans , SARS-CoV-2
13.
Cancer Immunol Immunother ; 59(5): 779-87, 2010 May.
Article in English | MEDLINE | ID: mdl-20091029

ABSTRACT

Adoptive transfer of antigen-specific cytotoxic T lymphocyte (CTL) into patients holds promise in treating cancer. Such anti-cancer CTL are stimulated by professional antigen-presenting dendritic cells (DC). We hypothesize the gene delivery of various Th1-response cytokines, such as interleukin 7 (IL-7), should further enhance CTL stimulation and activity. However, the issue as to which cell type, DC (paracrine) or the T cell (autocrine), should express a particular Th1 cytokine gene for optimal CTL stimulation has never been addressed. We used adeno-associated virus-2 (AAV) to compare delivery of IL-7 and IL-2 genes into DC or T cells and to exogenous commercial cytokines for generating robust carcinoembryonic antigen (CEA)-specific CTL. AAV/IL-7 transduction of T cells (autocrine delivery) generated CTL with the highest killing capability. Consistent with this, AAV/IL-7 delivery generated T cell populations with the highest proliferation, highest interferon gamma expression, highest CD8(+):CD4(+) ratio, highest CD8(+), CD69(+) levels, and lowest CD4(+), CD25(+) (Treg) levels. These data are consistent with higher killing by the AAV/IL-7-altered CTL. These data strongly suggest that IL-7 autocrine gene delivery is optimal for CTL generation. These data also suggest Th1 cytokine autocrine versus paracrine delivery is an important issue for immuno-gene therapy and uncovers new questions into cytokine mechanism of action.


Subject(s)
Dendritic Cells/immunology , Genetic Therapy/methods , Immunotherapy/methods , Interleukin-7/genetics , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes/immunology , Carcinoembryonic Antigen/genetics , Carcinoembryonic Antigen/immunology , Cell Separation , Dependovirus , Flow Cytometry , Genetic Vectors , Humans , Interleukin-2/genetics , Interleukin-7/immunology , T-Lymphocyte Subsets/immunology , Transduction, Genetic
14.
Breast Cancer Res Treat ; 122(1): 259-65, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20033766

ABSTRACT

Epidemiological studies have evaluated the association between CYP17 MspA1 polymorphism and breast cancer risk. However, the results remain conflicting rather than conclusive. To derive a more precise estimation of the relationship, we performed this meta-analysis. Systematic searches of the PubMed and Medline databases were performed. A total of 35 studies including 22,090 cases and 28,498 controls were identified. Genotype distributions of CYP17 in the controls of all studies were in agreement with Hardy-Weinberg equilibrium (HWE) except for three studies. When all 35 studies were pooled into the meta-analysis, there was no evidence for significant association between CYP17 MspA1 polymorphism and breast cancer risk (for A1/A2 vs. A1/A1: OR = 1.00, 95% CI = 0.96-1.04; for A2/A2 vs. A1/A1: OR = 1.03, 95% CI = 0.97-1.08; for dominant model: OR = 1.01, 95% CI = 0.97-1.05; for recessive model: OR = 1.03, 95% CI = 0.98-1.08). In the subgroup analyses by ethnicity, menopausal status and source of controls, no significant associations were found in all genetic models. When sensitivity analyses were performed by excluding HWE-violating studies, all the results were not materially altered. In summary, the meta-analysis strongly suggests that CYP17 MspA1 polymorphism is not associated with increased breast cancer risk.


Subject(s)
Breast Neoplasms/epidemiology , Polymorphism, Restriction Fragment Length , Polymorphism, Single Nucleotide , Steroid 17-alpha-Hydroxylase/genetics , Amino Acid Substitution , Breast Neoplasms/ethnology , Breast Neoplasms/genetics , Case-Control Studies , Ethnicity/genetics , Ethnicity/statistics & numerical data , Female , Genetic Predisposition to Disease , Genotype , Humans , Menopause , Models, Genetic , Odds Ratio , Patient Selection , Risk
15.
J Biomed Biotechnol ; 2010: 270985, 2010.
Article in English | MEDLINE | ID: mdl-20490265

ABSTRACT

The adoptive transfer of antigen-specific cytotoxic T lymphocytes (CTL) shows promise in the treatment of cancer and infectious diseases. We utilize adeno-associated virus-(AAV-) based antigen gene-loaded dendritic cells (DCs) to stimulate such antigen-specific CTL. Yet further improvements in CTL stimulation and killing may result by gene delivery of various Th1-response interferons/cytokines, such as interferon gamma (IFN-gamma), as the delivered gene can continuously produce that interferon. However which immune cell type should optimally express IFN-gamma is unclear as the phenotypes of both DC and T cells are enhanced by it. Here, we used AAV to compare and contrast IFN-gamma gene delivery into DC or T cells, and versus the addition of exogenous IFN-gamma, for stimulating carcinoembryonic antigen-(CEA-) specific CTL. It was found that AAV/IFN-gamma delivery into T cells (autocrine) resulted in T cell populations with the highest CD8(+)/CD4(+) ratio, highest IFN-gamma(+)/IL-4(+) ratio, highest CD69(+),CD8(+) levels, and lowest CD4(+)/CD25(+) levels, all consistent with the strongest Th1 response. Most importantly, AAV/IFN-gamma transduction of T cells resulted in antigen-specific T cell populations with the highest killing capabilities, 49% above other treatments. These data strongly suggest that AAV/IFN-gamma autocrine gene delivery into T cells is worthy of further study towards maximizing the generation of antigen-specific anticancer CTL killers.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Interferon-gamma/genetics , T-Lymphocytes, Cytotoxic/immunology , Carcinoembryonic Antigen/genetics , Carcinoembryonic Antigen/immunology , Cell Line, Tumor , Cell Proliferation , Cytokines/immunology , Dendritic Cells/immunology , Dependovirus/genetics , Epitopes, T-Lymphocyte/genetics , Flow Cytometry , Gene Transfer Techniques , Humans , Interferon-gamma/immunology , Lymphocyte Activation , Reverse Transcriptase Polymerase Chain Reaction , Virus Integration
16.
Zhonghua Zhong Liu Za Zhi ; 32(8): 575-9, 2010 Aug.
Article in Zh | MEDLINE | ID: mdl-21122407

ABSTRACT

OBJECTIVE: To establish a cetuximab-resistant human nasopharyngeal carcinoma 5-8F/Erbitux cell line and preliminarily study the relationship between the insulin-like growth factor 1 receptor (IGF-1R) signaling pathway and the resistance of nasopharyngeal carcinoma to cetuximab. METHODS: A nasopharyngeal cancer cell line, 5-8F, with high epidermal growth factor receptor (EGFR) expression and cetuximab sensitivity, was selected as study object. The cetuximab-resistant 5-8F/Erbitux cell line was induced by stepwise selection after exposure to increasing doses of cetuximab. The IC(50) was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and the resistance index (RI) was calculated. The growth curves of 5-8F and 5-8F/Erbitux cells were plotted and the doubling times were calculated by cell counting assay. The cell cycle was detected by flow cytometry. Cross-resistance profiles of 5-8F/Erbitux cells to 5-Fu, Taxol and DDP were tested by MTT assay. Expression levels of P-gP, IGF-1R and P-IGF-1R of 5-8F and 5-8F/Erbitux cells were determined by Western blot analysis and MDR1 gene by real-time fluorescent quantitative PCR. RESULTS: A cetuximab-resistant human nasopharyngeal carcinoma cell line 5-8F/Erbitux was successfully established and their resistance index (RI) were 1.2 and 1.1, respectively, at 3 d and 5 d of the cetuximab treatment. The doubling times of 5-8F and 5-8F/Erbitux cells were 26.63 h and 142.30 h, respectively. Flow cytometry demonstrated that 5-8F/Erbitux cells showed an increased population at G(0)/G(1) phase (P < 0.001) and reduced population at S phase (P < 0.001), compared with 5-8F cells. The 5-8F/Erbitux cells showed cross-resistance to 5-Fu (RI = 3.95, P < 0.01) and some resistance to Taxol as well as enhanced sensitivity to DDP (P > 0.05 for all). The 5-8F/Erbitux cells also had increased levels of P-gP, IGF-1R and P-IGF-1R compared with 5-8F cells (P < 0.001 for all). Expression of MDR1 gene was not detected in 5-8F cells and only very weak expression in 5-8F/Erbitux cells. CONCLUSION: Cetuximab-resistant 5-8F/Erbitux cells have no common multidrug resistance like that induced by traditional chemotherapeutic drugs. The excessive activation of the IGF-1R signaling pathway is probably one of the mechanisms that caused resistance of 5-8F/Erbitux cells to cetuximab.


Subject(s)
Antibodies, Monoclonal/pharmacology , Drug Resistance, Neoplasm , Nasopharyngeal Neoplasms/pathology , Receptor, IGF Type 1/metabolism , Signal Transduction , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antibodies, Monoclonal, Humanized , Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cetuximab , Cisplatin/pharmacology , Fluorouracil/pharmacology , Humans , Nasopharyngeal Neoplasms/metabolism , Paclitaxel/pharmacology , Phosphorylation
17.
Signal Transduct Target Ther ; 5(1): 13, 2020 02 14.
Article in English | MEDLINE | ID: mdl-32296025

ABSTRACT

MYH9 has dual functions in tumors. However, its role in inducing tumor stemness in hepatocellular carcinoma (HCC) is not yet determined. Here, we found that MYH9 is an effective promoter of tumor stemness that facilitates hepatocellular carcinoma pathogenesis. Importantly, targeting MYH9 remarkably improved the survival of hepatocellular carcinoma-bearing mice and promoted sorafenib sensitivity of hepatocellular carcinoma cells in vivo. Mechanistic analysis suggested that MYH9 interacted with GSK3ß and reduced its protein expression by ubiquitin-mediated degradation, which therefore dysregulated the ß-catenin destruction complex and induced the downstream tumor stemness phenotype, epithelial-mesenchymal transition, and c-Jun signaling in HCC. C-Jun transcriptionally stimulated MYH9 expression and formed an MYH9/GSK3ß/ß-catenin/c-Jun feedback loop. X protein is a hepatitis B virus (HBV)-encoded key oncogenic protein that promotes HCC pathogenesis. Interestingly, we observed that HBV X protein (HBX) interacted with MYH9 and induced its expression by modulating GSK3ß/ß-catenin/c-Jun signaling. Targeting MYH9 blocked HBX-induced GSK3ß ubiquitination to activate the ß-catenin destruction complex and suppressed cancer stemness and EMT. Based on TCGA database analysis, MYH9 was found to be elevated and conferred poor prognosis for hepatocellular carcinoma patients. In clinical samples, high MYH9 expression levels predicted poor prognosis of hepatocellular carcinoma patients. These findings identify the suppression of MYH9 as an alternative approach for the effective eradication of CSC properties to inhibit cancer migration, invasion, growth, and sorafenib resistance in HCC patients. Our study demonstrated that MYH9 is a crucial therapeutic target in HCC.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Glycogen Synthase Kinase 3 beta/genetics , Liver Neoplasms/drug therapy , Myosin Heavy Chains/genetics , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/drug effects , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing/drug effects , Heterografts , Humans , JNK Mitogen-Activated Protein Kinases/genetics , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Neoplastic Stem Cells/drug effects , Sorafenib/pharmacology , Trans-Activators/genetics , Ubiquitination/drug effects , Viral Regulatory and Accessory Proteins/genetics , Wnt Signaling Pathway/drug effects , beta Catenin/genetics
18.
Lasers Med Sci ; 24(4): 597-603, 2009 Jul.
Article in English | MEDLINE | ID: mdl-18936869

ABSTRACT

Our objective was to examine the effect of time intervals between Photofrin injection and laser irradiation [i.e., drug-light interval (DLI)] on the mode of action of Photofrin photodynamic therapy (PDT). Kunming mice transplanted with sarcoma-180 cells were used as an animal model. The tumor-bearing mice in the control group were given neither photosensitizer nor laser irradiation. PDT groups were given intravenous (i.v.) injection of Photofrin (7.5 mg/kg) prior to being irradiated with a 630 nm laser at 120 J/cm(2) at different DLIs (1 min-48 h). Tumors and overlying skin were visually examined daily. Histopathological and electron microscopic examinations were carried out 48 h after PDT. Survival rates were recorded. The mice in the groups that had experienced short DLIs (<60 min) showed stronger skin reactions than the groups subjected to long DLIs (>6 h). Histological examination showed that antitumor effects were achieved mainly by the destruction of tumor blood vessels and the formation of thrombosis at short DLIs, whereas, at long DLIs, the tumor cells were killed directly by PDT-mediated cytotoxicity. Electron microscopy revealed various degrees of mitochondrial swelling. The survival rate of the mice subjected to long DLIs was slightly higher than that of the mice subjected to short DLIs. Both vascular (e.g., tumor vessel destruction) and cellular (e.g., cytotoxicity) effects contributed to Photofrin PDT-induced tumor ablation.


Subject(s)
Dihematoporphyrin Ether/therapeutic use , Photochemotherapy/methods , Photosensitizing Agents/therapeutic use , Sarcoma 180/drug therapy , Animals , Dihematoporphyrin Ether/administration & dosage , Lasers, Semiconductor , Low-Level Light Therapy , Male , Mice , Microscopy, Electron, Transmission , Neoplasm Transplantation , Photochemotherapy/adverse effects , Photosensitizing Agents/administration & dosage , Sarcoma 180/blood supply , Sarcoma 180/pathology , Skin/pathology , Skin/radiation effects , Time Factors
19.
Int J Clin Exp Pathol ; 11(4): 2092-2099, 2018.
Article in English | MEDLINE | ID: mdl-31938317

ABSTRACT

Cancer stem cells (CSCs) are responsible for cancer recurrence and metastasis and are related to poor prognosis in patients with hepatocellular carcinoma (HCC). CD133 is one of the most commonly used CSC markers. In this study, expression and the biological significance of CSC marker CD133 was evaluated in HCC, at mRNA and protein levels. We demonstrate that both mRNA and protein levels of CD133 are significantly elevated in HCC relative to that in adjacent non-cancerous tissue based on bioinformatics and immunohistochemical analysis, respectively (P < 0.01). Intriguingly, we detected nuclear distribution of CD133 and found that nuclear CD133 expression was indicative of poor patient prognosis (median survival 12 months versus 34.5 months) (Log-Rank, P = 0.0258). Meanwhile, our findings suggest that nuclear CD133 expression is positively correlated with tumor size and serves as an independent prognostic factor for HCC after surgical resection (HR = 0.564, 95% CI 0.313-1.018, P = 0.057). Nuclear CD133 expression can potentially serve as a biomarker for clinical diagnosis and prognosis of HCC.

20.
Int J Clin Exp Pathol ; 11(5): 2784-2791, 2018.
Article in English | MEDLINE | ID: mdl-31938396

ABSTRACT

Hepatocellular carcinoma (HCC) is currently one of the most common causes of cancer-related death and one of the most commonly diagnosed cancers. MYH9 is thought to play a critical role in cancer progression. However, the expression and prognostic value of MYH9 in HCC are still unknown. In this study, the expression and biological significance of MYH9 were evaluated in HCC, at mRNA and protein levels. We showed that both mRNA and protein levels of MYH9 were significantly upregulated in HCC relative to the levels in adjacent non-tumor tissues based on the TCGA database and immunohistochemical analysis, respectively (P < 0.001). Additionally, we found that high MYH9 protein levels correlated with poor patient prognosis (median survival 19 months versus 49 months) (Log-Rank, P = 0.0292). Meanwhile, our data suggested that MYH9 expression is an independent prognostic factor for HCC after surgical resection (HR = 0.675, 95% CI 0.452-1.008, P = 0.054) and can potentially serve as a biomarker for the clinical diagnosis and prognosis of HCC.

SELECTION OF CITATIONS
SEARCH DETAIL