Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Yoko Takagi is active.

Publication


Featured researches published by Yoko Takagi.


Clinical Cancer Research | 2004

Cyclooxygenase-2 Expression A Significant Prognostic Indicator for Patients With Colorectal Cancer

Labile Togba Soumaoro; Hiroyuki Uetake; Tetsuro Higuchi; Yoko Takagi; Masayuki Enomoto; Kenichi Sugihara

Purpose: Recent studies have shown that cyclooxygenase (Cox)-2 may be involved in colorectal carcinogenesis. We aimed to determine whether Cox-2 expression in itself can predict outcome of colorectal cancer patient after surgery. In addition, the expression of Cox-1 was also evaluated. Experimental Design: Tissue samples of primary and secondary tumors from 288 patients undergoing surgical resections for colorectal adenocarcinoma were immunohistochemically examined for Cox-2 and Cox-1 expressions. The specimens were graded based on the intensity and extent of staining; then, the correlations between Cox-2 and Cox-1 expressions with clinicopathologic parameters and survival time were analyzed. Results: Expression of Cox-2 was positive in 70.8% of primary tumor, 92.0% of lymph node metastases, 100.0% of hepatic metastases, and was significantly associated with tumor size, depth of invasion, lymph node metastasis, vessels invasion, stage and recurrence. In contrast, Cox-1 was positive in 42.7% of primary tumor, 84.0% of lymph node metastases, 37.5% hepatic metastases, and was associated with only tumor size. Patients with Cox-2–positive tumors had a significant shorter survival time than those with negative tumors did (P = 0.0006 by log-rank test); and, in a multivariate analysis, Cox-2 was an independent prognostic factor (P = 0.0103; relative risk 4.114; 95% confidence interval, 1.397–12.120). Cox-1 status had no statistically effect on patient survival time. Conclusions: Elevated Cox-2 expression, but not that of Cox-1, was significantly associated with reduced survival and recognized as an independent prognostic factor in our cohort of colorectal cancer patients.


Annals of Surgical Oncology | 2008

Lactate Dehydrogenase-5 (LDH-5) Expression in Human Gastric Cancer: Association with Hypoxia-Inducible Factor (HIF-1α) Pathway, Angiogenic Factors Production and Poor Prognosis

Yanislav Kolev; Hiroyuki Uetake; Yoko Takagi; Kenichi Sugihara

BackgroundLactate-dehydrogenase-5 (LDH-5) is an important isoenzyme converting pyruvate to lactate under hypoxic conditions and might play an important role in the development and progression of malignancies. However, the role of LDH-5 in gastric cancer is still unclear. In this study, we investigated the clinical significance of LDH-5 expression in gastric carcinoma.MethodsLDH-5 expression in 152 patients with different grade and stage gastric carcinoma was analyzed by immunohistochemistry. In addition, hypoxia-inducible factor 1α (HIF-1α) as a marker of tumor hypoxia, as well as vascular endothelial growth factor (VEGF) and cyclooxygenase-2 (COX-2) as angiogenesis parameters were also assessed in this study. Correlations between the expression of investigated proteins and various clinicopathological factors including survival were determined.ResultsThere were 94 cases (61.8%) showing high LDH-5 expression, and 95 patients (62.5%) had high HIF-1α expression. Positive correlation was found between LDH-5 expression and HIF-1α, VEGF, and COX-2. The overexpression of LDH-5 was more prevalent in advanced tumors having positive vessel invasion. Patients with overexpression of LDH-5 showed far lower disease-free (63.5% vs 82.7%) and overall (56.3% vs 78.4%) survival rates compared with patients with low LDH-5 expression. HIF-1α expression was shown to have no significance on survival. In multivariate analysis, high LDH-5 expression kept its independence as a negative prognostic indicator.ConclusionThe results of the current study show that LDH-5 expression may be a useful prognostic factor for patients with gastric carcinoma.


International Journal of Cancer | 2007

PIK3CA mutation is predictive of poor survival in patients with colorectal cancer

Shunsuke Kato; Satoru Iida; Tetsuro Higuchi; Toshiaki Ishikawa; Yoko Takagi; Masamichi Yasuno; Masayuki Enomoto; Hiroyuki Uetake; Kenichi Sugihara

The PI3K‐AKT pathway is activated in a variety of human cancers, resulting in disturbance of cell growth, proliferation and survival. Among the factors affecting the pathway, the K‐Ras mutation and PIK3CA mutation are the most common oncogenic alterations in colorectal cancer. We hypothesized that these two mutations are important in activation of the PI3K pathway and colorectal carcinogenesis. In this study, we aimed to examine the influence of PIK3CA mutation and K‐Ras mutation on AKT activation, and to clarify whether PIK3CA mutation, K‐Ras mutation and p‐AKT expression may be used as parameters for predicting prognosis in colorectal cancer. Tissue samples from 158 colorectal cancer patients who underwent surgical resection were examined. The sequences of exon 1 of K‐Ras and exons 9 and 20 of PIK3CA were determined by direct sequencing using genomic DNA extracted from paraffin‐embedded blocks. Activation status of AKT was evaluated by immunohistochemical staining using phospho‐specific AKT antibody (Ser473). Correlation between these factors and clinicopathologic findings/patient survival were examined. As a result, PIK3CA mutation was significantly associated with shorter relapse‐free survival (RFS) in stage II/III patients (p = 0.0216) and shorter disease‐specific survival in all patients (p = 0.0357). In the multivariate analysis, PIK3CA mutation was the only independent and significant prognostic factor for RFS in stage II/III patients (p = 0.0433, HR 2.478). This study revealed the prognostic value of PIK3CA mutation in colorectal cancer patients. Patients with PIK3CA mutation should be followed up carefully. Moreover, our result suggests that inhibition of PIK3CA mutant may be a new molecular target therapy.


Clinical Cancer Research | 2008

High Expression of HER3 Is Associated with a Decreased Survival in Gastric Cancer

Mikiko Hayashi; Mikito Inokuchi; Yoko Takagi; Hiroyuki Yamada; Kazuyuki Kojima; Jiro Kumagai; Tatsuyuki Kawano; Kenichi Sugihara

Background: The role of human epidermal growth factor receptor (HER) 3 and HER4 has been elucidated in gastric cancer. HER1 and HER2 overexpression are regarded as prognostic factors and targets of treatment. The dimerization of the HER family receptors activates downstream signal pathways and promotes tumor progression. This study investigated the positive correlation between HER1 and HER4 expression and the prognosis of patients with gastric cancers. Experimental Design: Tumor samples were obtained from gastric adenocarcinomas of 134 patients who underwent a gastrectomy from 1999 to 2002. The expression of each HER was analyzed in the tumor by immunohistochemical staining. Parametric correlations were done between HER expression and the clinicopathologic findings. A multivariate analysis was done with the overall survival. Results: HER3 expression was significantly associated with parameters involved with tumor progression, including the depth of tumor invasion (T1 versus T2-T4; P = 0.000), involved lymph nodes (P = 0.000), distant metastasis (P = 0.008), tumor stage (P = 0.000), and recurrent disease (P = 0.000). HER1 was also significantly associated with those factors excluding distant metastasis. A significant relationship was observed between the expression of HER1 and HER3 (P = 0.000). HER3 overexpression was associated with a significantly worse survival (P = 0.0000) and was an independent prognostic factor in the multivariate analysis (hazard ratio, 2.382; 95% confidence interval, 1.009-5.625; P = 0.048). Conclusions: HER3 overexpression is strongly associated with tumor progression and poor prognosis of patients with gastric cancer. It may become a new prognostic factor and a target of treatment.


International Journal of Cancer | 2008

Methylated TMS1 and DAPK genes predict prognosis and response to chemotherapy in gastric cancer.

Keiji Kato; Satoru Iida; Hiroyuki Uetake; Yoko Takagi; Toshiki Yamashita; Mikito Inokuchi; Hiroyuki Yamada; Kazuyuki Kojima; Kenichi Sugihara

Gastric cancer is the second most common cause of cancer deaths worldwide. The identification of molecular genetic parameters that are associated with response to chemotherapy and prognosis is of utmost interest. We examined methylation of the apoptosis‐related genes, TMS1 and DAPK, in 81 primary gastric cancers using methylation‐specific PCR and compared their methylation status with clinicopathological findings. Aberrant methylation of TMS1 and DAPK genes was detected in 26 (32.1%) tumors and in 18 (22.2%) tumors, respectively. The overall survival of patients with both methylated genes was significantly shorter compared with those with only one methylated gene or no methylated genes (p = 0.0003). Neither gene methylation had any relation to other clinicopathological findings. Next, we examined 43 patients treated by 5‐fluorouracil–based chemotherapy, who had distant metastasis or recurrence after radical resection, to determine the relation between chemosensitivity and methylation. The response rate was lower in patients with either methylation than without (TMS1: 22.2% vs. 48.0%; DAPK: 21.4% vs. 44.8%). Overall survival tended to be shorter in the patients with both methylations compared with either or no methylations (p = 0.0806). The time to progression of patients with methylation of TMS1 or DAPK was significantly shorter than patients without methylation (TMS1: p = 0.0123; DAPK: p = 0.0464). Furthermore, the time to progression of patients with both methylated genes was significantly shorter than patients with one methylation or no methylation (p = 0.0082). In conclusion, TMS1 and DAPK methylation might predict the prognosis and response to chemotherapy in gastric cancer.


Oncology Reports | 2011

Methylation of BNIP3 and DAPK indicates lower response to chemotherapy and poor prognosis in gastric cancer

Hirofumi Sugita; Satoru Iida; Mikito Inokuchi; Keiji Kato; Megumi Ishiguro; Toshiaki Ishikawa; Yoko Takagi; Megumu Enjoji; Hiroyuki Yamada; Hiroyuki Uetake; Kazuyuki Kojima; Kenichi Sugihara

Aberrant promoter hypermethylation (methylation) is an epigenetic change that silences the expression of crucial genes, thus inactivating the apoptotic pathway in various cancers. Inactivation of the apoptotic pathway has been considered to be associated with chemoresistance. The objective of the present study was to clarify the effect of the methylation of the apoptosis-related genes, Bcl-2/adenovirus E1B 19 kDa-interacting protein 3 (BNIP3) and death-associated protein kinase (DAPK), on the response to chemotherapy in metastatic or recurrent gastric cancers. Tumor samples were obtained from 80 gastric cancer patients who were treated with fluoropyrimidine-based chemotherapy for distant metastatic or recurrent disease, after surgical resection of the primary tumor. The methylation status of the apoptosis-related genes, BNIP3 and DAPK, was investigated by methylation-specific PCR. Methylation in BNIP3 was detected in 31 tumors (39%) and in DAPK in 33 tumors (41%). There was no correlation between the methylation status of BNIP3 and that of DAPK. The response rate was significantly lower in patients with methylation of DAPK, than in those without (21 vs. 49% p=0.012). Progression-free survival time (PFS) was shorter in patients with methylation of DAPK than in those without (p=0.007). The overall survival time (OS) was shorter in patients with methylation of BNIP3 than in those without (p=0.031). The response rate was significantly lower in patients with methylation of either DAPK or BNIP3, or both, than in those without methylation (p=0.003). PFS and OS were significantly shorter in patients with methylation of either or both of these genes than in those without (p=0.002, p=0.001). The methylation of BNIP3 and DAPK can predict lower response to chemotherapy and poor prognosis in gastric cancer.


Oncology Reports | 2011

Vimentin expression is associated with decreased survival in gastric cancer.

Sho Otsuki; Mikito Inokuchi; Megumu Enjoji; Toshiaki Ishikawa; Yoko Takagi; Keiji Kato; Hiroyuki Yamada; Kazuyuki Kojima; Kenichi Sugihara

Gastric cancer rich in stromal tissue, such as diffuse-type disease, carries a poor prognosis. In some cancers, expression of vimentin, a mesenchymal maker, is associated with poor survival. The expression of mesenchymal markers such as vimentin is observed after epithelial-mesenchymal transition (EMT), an important initial behavioral change related to the adhesion and migration properties of tumor cells that is required for local tumor invasion. A hallmark of EMT is the loss of E-cadherin. EMT-inducing regulators, including SIP1, Slug, and Twist, repress E-cadherin transcription by interacting with E-cadherin promoter. We investigated the expression of vimentin and EMT-related genes, including SIP1, Slug, and Twist, in frozen cancer tissues and normal tissues by real-time quantitative reverse-transcriptase polymerase chain reaction. Tumor samples were obtained from 106 patients with gastric adenocarcinomas who underwent a gastrectomy. The relation of the expression of these genes to clinicopathological factors and outcomes was studied. Vimentin mRNA was significantly higher in diffuse type compared to intestinal type according to Laurens classification (p=0.048) and was significantly elevated in patients with recurrent or distant metastatic disease (p=0.049). Immunohistochemically, however, vimentin was detected only in cancer stroma. Twist mRNA expression significantly correlated with tumor depth (p=0.042) and advanced tumor stage (I-II vs. III-IV, p=0.030). E-cadherin immunohistochemical expression was significantly associated with Laurens histopathological type (p<0.001). Univariate analysis of relapse-free survival showed that tumor depth, lymph node metastasis, Laurens histopathological type, and vimentin mRNA expression were significant prognostic factors (p<0.001, p=0.013, p=0.011, and p=0.019). On multivariate analysis, vimentin mRNA expression was an independent prognostic factor [hazard ratio (HR)=2.1; 95% confidence interval (CI), 1.0-4.4; p=0.036], coming after tumor depth (HR=9.7; 95%CI, 3.7-24; p<0.001). Vimentin mRNA expression is associated with recurrence or distant metastasis and decreased survival in gastric cancer.


Oncology Reports | 2011

Effect of classification based on combination of mutation and methylation in colorectal cancer prognosis

Haruhiko Aoyagi; Satoru Iida; Hiroyuki Uetake; Toshiaki Ishikawa; Yoko Takagi; Hirotoshi Kobayashi; Tetsuro Higuchi; Masamichi Yasuno; Masayuki Enomoto; Kenichi Sugihara

Colorectal cancer (CRC) is caused by an accumulation of genetic alterations and epigenetic alterations. The molecular classification of CRCs based on genetic alterations and epigenetic alterations is evolving. Here, we examined mutations and methylation status in CRCs to determine if the combination of genetic and epigenetic alterations predicts prognosis. We examined 134 sporadic CRCs. We used the direct sequencing method to identify mutations in BRAF and AKT1, which are downstream of KRAS and PIK3CA, respectively, in the EGFR pathway. We used the Methylight method to determine the methylation status of hMLH1, p16, MINT1, MINT2 and MINT31. Both BRAF and AKT1 mutations were found in only one case (0.75%). Aberrant methylation of hMLH1, p16, MINT1, MINT2 and MINT31 was detected in 22.4, 35.1, 32.8, 59.7 and 41.0% of cases, respectively. The clinicopathological factors were not significantly correlated to mutation or methylation. Among the patients who had no mutation in the EGFR pathway, the overall survival was significantly shorter in the patients with methylation compared to the patients with no methylation in hMLH1 and p16 (p=0.0318). Methylation could play a key role in the prognosis of patients without mutations in the EGFR pathway. The combination of genetic and epigenetic alterations may be a good marker for the prognosis of CRC patients.


Annals of Surgical Oncology | 2007

Effect of Combined Therapy With Low-Dose 5-Aza-2′-Deoxycytidine and Irinotecan on Colon Cancer Cell Line HCT-15

Megumi Ishiguro; Satoru Iida; Hiroyuki Uetake; Shinji Morita; Hiroshi Makino; Keiji Kato; Yoko Takagi; Masayuki Enomoto; Kenichi Sugihara

BackgroundAberrant promoter hypermethylation is an epigenetic change that silences the expression of crucial genes, resulting in inactivation of the apoptotic pathway in various cancers. This hypermethylation can be restored by the demethylating agent 5-aza-2′-deoxycytidine (DAC). DAC might increase the tumor sensitivity to chemotherapy through demethylation and restoration of gene expression. We investigated the effect of combined therapy with DAC and irinotecan (CPT-11) on the human colon cancer cell line HCT-15.MethodsHuman colon cancer cell line HCT-15 was treated with DAC and/or CPT-11 both in vitro and in vivo. The changes in mRNA expression of several apoptosis-related genes were investigated by reverse transcriptase–polymerase chain reaction (PCR). Promoter methylation was detected by methylation-specific PCR and combined bisulfite restriction analysis. Suppression of tumor growth was observed during the treatment with DAC and/or CPT-11 and apoptosis in the tumors was investigated by TUNEL (terminal deoxynucleotidyl transferase dUTP nick-end labeling) assay.ResultsPromoter methylation of p14ARF, p16INK4a, BNIP3, and XAF1 was confirmed, and DAC restored mRNA expression of these genes. Demethylation and restoration of gene expression was observed with low-dose DAC, and demethylation status was sustained for several weeks. Combined therapy with DAC and CPT-11 produced marked suppression in tumor growth compared with DAC or CPT-11 alone, both in vitro and in vivo.ConclusionsPretreatment with low-dose DAC may have the potential to be used as a “biosensitizer” of DNA-damaging agents such as CPT-11 when the apoptotic pathway is inactivated as a result of aberrant promoter methylation in the cancer.


Journal of Experimental & Clinical Cancer Research | 2012

Prognostic value of RKIP and p-ERK in gastric cancer

Yoshitaka Fujimori; Mikito Inokuchi; Yoko Takagi; Keiji Kato; Kazuyuki Kojima; Kenichi Sugihara

BackgroundThe mitogen-activated protein kinase (MAPK) signaling pathway participates in several steps of tumour development and is considered a prominent therapeutic target for the design of chemotherapeutic agents. We evaluated the expressions of extracellular signal-regulated kinase (ERK), mitogen-activated protein kinase (MEK), an upstream regulator of ERK, and Raf kinase inhibitor protein (RKIP), and investigated correlations of these expressions with clinicopathological features and outcomes in gastric cancer.MethodsTumour samples were obtained from 105 patients with gastric adenocarcinomas who underwent radical gastrectomy. The expressions of phosphorylated ERK (p-ERK), phosphorylated MEK (p-MEK), and RKIP were analysed by immunohistochemical staining.ResultsExpression of RKIP, p-MEK, and p-ERK was found in 69 (66%), 54 (51%), and 64 (61%) of all tumours, respectively. RKIP expression negatively correlated with the depth of invasion (p < 0.001), lymph node involvement (p = 0.028), and Union for International Cancer Control (UICC) stage (p = 0.007). RKIP expression was associated with significantly longer relapse-free survival (RFS) (p = 0.0033), whereas p-MEK was not (p = 0.79). Patients with p-ERK expression had slightly, but not significantly shorter RFS than those without such expression (p = 0.054). Patients with positive p-ERK and negative RKIP expression had significantly shorter RFS than the other patients (p < 0.001). The combination of RKIP and p-ERK expression was an independent prognostic factor (hazard ratio, 2.4; 95% confidence interval, 1.3 - 4.6; p = 0.008).ConclusionsOur results demonstrated that loss of RKIP was associated with tumour progression and poor survival. Negative RKIP expression combined with positive p-ERK expression was an independent predictor of poor outcomes in patients with gastric cancer.

Collaboration


Dive into the Yoko Takagi's collaboration.

Top Co-Authors

Avatar

Kenichi Sugihara

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Hiroyuki Uetake

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Kazuyuki Kojima

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Mikito Inokuchi

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Satoru Iida

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Keiji Kato

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Sho Otsuki

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Toshiaki Ishikawa

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Masayuki Enomoto

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Tatsuyuki Kawano

Tokyo Medical and Dental University

View shared research outputs
Researchain Logo
Decentralizing Knowledge