Network


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

Hotspot


Dive into the research topics where Hideki Terai is active.

Publication


Featured researches published by Hideki Terai.


Molecular Cancer Research | 2013

Activation of the FGF2-FGFR1 Autocrine Pathway: A Novel Mechanism of Acquired Resistance to Gefitinib in NSCLC

Hideki Terai; Kenzo Soejima; Hiroyuki Yasuda; Sohei Nakayama; Junko Hamamoto; Daisuke Arai; Kota Ishioka; Keiko Ohgino; Shinnosuke Ikemura; Takashi Sato; Satoshi Yoda; Ryosuke Satomi; Katsuhiko Naoki; Tomoko Betsuyaku

Patients with non-small cell lung cancer (NSCLC) that harbors epidermal growth factor receptor (EGFR) mutations initially respond to EGFR-tyrosine kinase inhibitors (TKI) but eventually experience relapse. Acquired resistance to EGFR-TKIs is strongly associated with patient mortality. Thus, elucidation of the mechanism of acquired resistance to EGFR-TKIs is of great importance. In this study, gefitinib-resistant cell line models were established by long-term exposure to gefitinib using the gefitinib-sensitive lung cancer cell lines, PC9 and HCC827. Expression analyses indicated that both FGFR1 and FGF2 were increased in PC9 gefitinib-resistant (PC9 GR) cells as compared with PC9 naïve (PC9 na) cells. Importantly, proliferation of gefitinib-resistant cells was dependent on the FGF2 -FGFR1 pathway. Mechanistically, inhibition of either FGF2 or FGFR1 by siRNA or FGFR inhibitor (PD173074) restored gefitinib sensitivity in PC9 GR cells. These data suggest that FGF2 -FGFR1 activation through an autocrine loop is a novel mechanism of acquired resistance to EGFR-TKIs. Mol Cancer Res; 11(7); 759–67. ©2013 AACR.


Molecular Medicine Reports | 2013

Identification of microRNAs differentially expressed between lung squamous cell carcinoma and lung adenocarcinoma

Junko Hamamoto; Kenzo Soejima; Satoshi Yoda; Katsuhiko Naoki; Sohei Nakayama; Ryosuke Satomi; Hideki Terai; Shinnosuke Ikemura; Takashi Sato; Hiroyuki Yasuda; Yuichiro Hayashi; Michiie Sakamoto; Toru Takebayashi; Tomoko Betsuyaku

Recent advances in the treatment of non-small cell lung cancer (NSCLC) with new agents require accurate histological subtyping at diagnosis to avoid the higher risk of an adverse response and to obtain the maximum therapeutic response. However, interobserver variability, tumor heterogeneity and the degree of differentiation may affect the decision concerning a pathological diagnosis of NSCLC. Therefore, the aim of this study was to identify specific microRNAs (miRNAs) as standardized biomarkers with high sensitivity and specificity in order to distinguish between squamous cell carcinoma (SCC) and adenocarcinoma (AC). Quantitative polymerase chain reaction (qPCR)‑based miRNA array analysis was performed to identify microRNAs differentially expressed between SCC and AC using 86 resected NSCLC samples in addition to adjacent normal tissues. The results were confirmed by independent qRT-PCR assays with the same test samples and 88 additional validation samples, and from this we evaluated the usefulness of the identified miRNAs as biomarkers to distinguish between SCC and AC. Three miRNAs (hsa-miR-196b, hsa-miR-205 and hsa-miR-375) were identified. Discriminant analysis combining the three miRNAs appeared to distinguish SCC from AC accurately in the test and validation samples, demonstrating a sensitivity and specificity of 76 and 80%, and 85 and 83%, respectively. hsa-miR-196b, hsa-miR-205 and hsa-miR-375 were identified as biomarkers capable of distinguishing between lung SCC and lung AC. These newly identified miRNAs may prove to be highly valuable molecular markers for the classification of NSCLC histological subtypes and may contribute to the pathogenesis of each subtype of NSCLC.


Molecular Cancer Research | 2010

The Combination of Multiple Receptor Tyrosine Kinase Inhibitor and Mammalian Target of Rapamycin Inhibitor Overcomes Erlotinib Resistance in Lung Cancer Cell Lines through c-Met Inhibition

Ichiro Nakachi; Katsuhiko Naoki; Kenzo Soejima; Ichiro Kawada; Hideo Watanabe; Hiroyuki Yasuda; Sohei Nakayama; Satoshi Yoda; Ryosuke Satomi; Shinnosuke Ikemura; Hideki Terai; Takashi Sato; Akitoshi Ishizaka

Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKI) show antitumor activity in a subset of non–small cell lung cancer (NSCLC) patients. However, the initial tumor response is followed by recurrence. Several studies have suggested the importance of other receptor tyrosine kinases (RTK) and downstream kinases as potential targets in the treatment of NSCLC. We used the multiple-RTK inhibitor AEE788, which inhibits EGFR, vascular endothelial growth factor receptor, and human epidermal growth factor receptor 2, with and without the downstream kinase inhibitor RAD001 (an inhibitor of mammalian target of rapamycin). AEE788 inhibited cell growth more effectively than did erlotinib in three NSCLC cell lines examined (A549, H1650, and H1975). However, in the EGFR-TKI–resistant cell line H1975 harboring T790M resistance mutation, cell growth inhibition by AEE788 was only mild, and the phosphorylation of its leading targets such as EGFR and vascular endothelial growth factor receptor 2 was not inhibited. In H1975, AEE788 induced significantly greater cell growth inhibition when combined with RAD001 than when used alone. This cooperative effect was not seen with the combination of erlotinib and RAD001. We found that c-Met was highly phosphorylated in this cell line, and the phosphorylated c-Met was inhibited effectively by AEE788. Using a phospho-RTK array, the phosphorylation of c-Met and insulin-like growth factor-I receptor was inhibited by AEE788. These results suggest that upstream RTK inhibitor overcomes the acquired resistance to EGFR-TKI when combined with downstream kinase inhibitor. Thus, the combined inhibition of upstream and downstream RTKs is a promising strategy for the treatment of NSCLC. Mol Cancer Res; 8(8); 1142–51. ©2010 AACR.


Lung Cancer | 2011

Bronchoscopic Microsampling is a Useful Complementary Diagnostic Tool for Detecting Lung Cancer

Hiroyuki Yasuda; Kenzo Soejima; Sohei Nakayama; Ichiro Kawada; Ichiro Nakachi; Satoshi Yoda; Ryosuke Satomi; Shinnosuke Ikemura; Hideki Terai; Takashi Sato; Hideo Watanabe; Katsuhiko Naoki; Yuichiro Hayashi; Akitoshi Ishizaka

PURPOSE Bronchoscopic microsampling (BMS) is a novel and direct method with which to obtain epithelial lining fluid (ELF) from the lungs. Analysis of DNA hypermethylation of tumor suppressor genes (TSGs) is expected to be a sensitive tool for the early detection of lung cancer. It has been reported that the existence of EGFR mutations and EML4-ALK gene rearrangements are related to the sensitivity of corresponding kinase inhibitors. We aimed to evaluate the suitability of ELF as a sample for analyzing molecular changes specific for lung cancer. PATIENTS AND METHODS We collected ELF from 61 lung cancer patients by BMS from the airway close to the peripheral lung nodule and purified the nucleic acids. We performed methylation specific PCR in each ELF as well as matched serum and tumor tissue for TSGs for DNA methylation analysis. We also examined EGFR mutations and EML4-ALK rearrangement. RESULTS The sensitivity for detecting DNA hypermethylation in ELF vs serum was 74.1% vs 18.5%. We found 60.1% of patients had at least one hypermethylation in ELF, while only 27.9% had it in serum. Of note, DNA hypermethylation was detected even in stage I patients (60.0%) and the detection rate was almost the same level in each stage. We also found the sensitivity for detecting EGFR mutation in ELF vs serum was 58.3% vs 8.3%. We detected an EML4-ALK fusion gene using ELF in one patient. CONCLUSIONS BMS is an alternative method to detect cancer specific genetic and epigenetic alterations and will be a useful complementary diagnostic tool for lung cancer. SUMMARY Investigation of genetic and epigenetic changes associated with lung cancer has clinical importance for its diagnosis and management. The clinical usefulness of bronchoscopic microsampling (BMS) in lung cancer has not yet been evaluated. This study demonstrates that BMS could be useful for detecting lung cancer specific molecular changes and valuable for early diagnosis and determination of treatment options for lung cancer.


International Journal of Chronic Obstructive Pulmonary Disease | 2015

The reasons for triple therapy in stable COPD patients in Japanese clinical practice

Masaki Miyazaki; Hidetoshi Nakamura; Saeko Takahashi; Shotaro Chubachi; Mamoru Sasaki; Mizuha Haraguchi; Hideki Terai; Koichi Fukunaga; Sadatomo Tasaka; Kenzo Soejima; Koichiro Asano; Tomoko Betsuyaku

Background Triple combination therapy involving long-acting muscarinic antagonists long-acting β2 agonists, and inhaled corticosteroids has recently become an option for maintenance treatment of COPD. Some add-on clinical trials have reported the benefits of these combinations. However, the process to step up to triple therapy varies for individual cases. Methods Keio University and affiliated hospitals conducted an observational COPD cohort study, recruiting patients diagnosed as having COPD by pulmonary physicians and those referred for investigation of possible COPD. Their prescription history and clinical course were retrospectively analyzed based on the physicians’ medical records and patient questionnaires. This study was registered with UMIN (UMIN000003470, April 10, 2010). Results A total of 95 of the 445 COPD patients (21%) were treated with inhaled corticosteroids/long-acting β2 agonists/long-acting muscarinic antagonists as maintenance therapy, including 12 in COPD Grade I, 31 in Grade II, 38 in Grade III, and 14 in Grade IV, based on the Global Initiative for Chronic Obstructive Lung Disease spirometric grading. For more than half of the patients on triple therapy, the treatment had been intensified due to unsatisfactory improvement of symptoms, and 32% were treated with triple therapy due to comorbid asthma. In contrast, there were COPD patients whose therapy was maintained after starting with triple therapy because of their serious conditions or concurrent exacerbation at diagnosis (8%). Conclusion Triple therapy was often prescribed in the real-life management of COPD, even in patients whose airflow limitation was not severe. To better control symptoms was the major reason for choosing triple therapy, regardless of the severity of COPD, in Japan.


Cancer Science | 2015

Methylation‐induced downregulation of TFPI‐2 causes TMPRSS4 overexpression and contributes to oncogenesis in a subset of non‐small‐cell lung carcinoma

Junko Hamamoto; Kenzo Soejima; Katsuhiko Naoki; Hiroyuki Yasuda; Yuichiro Hayashi; Satoshi Yoda; Sohei Nakayama; Ryosuke Satomi; Hideki Terai; Shinnosuke Ikemura; Takashi Sato; Daisuke Arai; Kota Ishioka; Keiko Ohgino; Tomoko Betsuyaku

We identified transmembrane protease, serine 4 (TMPRSS4) as a putative, druggable target by screening surgically resected samples from 90 Japanese non‐small‐cell lung cancer (NSCLC) patients using cDNA microarray. TMPRSS4 has two druggable domains and was upregulated in 94.5% of the lung cancer specimens. Interestingly, we found that TMPRSS4 expression was associated with tissue factor pathway inhibitor 2 (TFPI‐2) expression in these clinical samples. In contrast to TMPRSS4, TFPI‐2 expression was downregulated in NSCLC samples. The in vitro induction of TFPI‐2 in lung cancer cell lines decreased the expression of TMPRSS4 mRNA levels. Reporter assay showed that TFPI‐2 inhibited transcription of TMPRSS4, although partially. Knockdown of TMPRSS4 reduced the proliferation rate in several lung cancer cell lines. When lung cancer cell lines were treated with 5‐aza‐2′‐deoxycytidine or trichostatin A, their proliferation rate and TMPRSS4 mRNA expression levels were also reduced through the upregulation of TFPI‐2 by decreasing its methylation in vitro. The TFPI‐2 methylation level in the low TMPRSS4 group appeared to be significantly low in NSCLC samples (P = 0.02). We found a novel molecular mechanism that TFPI‐2 negatively regulates cell growth by inhibiting transcription of TMPRSS4. We suggest that TMPRSS4 is upregulated by silencing of TFPI‐2 through aberrant DNA methylation and contributes to oncogenesis in NSCLC.


Japanese Journal of Clinical Oncology | 2015

A Phase II study of S-1 and irinotecan combination therapy in previously treated patients with advanced non-small cell lung cancer

Shinnosuke Ikemura; Katsuhiko Naoki; Hiroyuki Yasuda; Ichiro Kawada; Satoshi Yoda; Hideki Terai; Takashi Sato; Kota Ishioka; Daisuke Arai; Keiko Ohgino; Hirofumi Kamata; Jun Miyata; Tomoko Betsuyaku; Kenzo Soejima

OBJECTIVE This Phase II study was conducted to evaluate the efficacy and safety of S-1 and irinotecan combination therapy as a second-line treatment in patients with advanced non-small cell lung cancer. METHODS Irinotecan was administered at 60 mg/m(2) on Days 1 and 8. Oral S-1 was administered on Days 1-14 every 3 weeks at 80 mg/day for patients with a body surface area of <1.25 m(2), 100 mg/day for patients with a body surface area of 1.25-1.5 m(2) and 120 mg/day for patients with a body surface area of >1.5 m(2). The primary endpoint was response rate, while the secondary endpoints were progression-free survival, overall survival and safety. RESULTS Thirty-one patients were enrolled in this study. The response and disease control rates were 6.5 and 58.1%, respectively. Progression-free survival and median survival time were 2.8 and 12.6 months, respectively. Grade 3-4 adverse events were reported for 29.0% of the patients. Hematological toxicities of Grade 3 or 4 included leukopenia (9.7%), neutropenia (9.7%), febrile neutropenia (3.2%), thrombopenia (3.2%) and anemia (6.5%). Non-hematological toxicities of Grade 3 or 4 included pneumonitis (6.5%), diarrhea, colitis, dyspnea, rash, oral mucositis, anorexia and pulmonary thromboembolism/deep vein thrombosis (3.2% each). CONCLUSIONS S-1 and irinotecan combination therapy at the present dose and schedule exhibited only modest efficacy with mild toxicities in previously treated patients with non-small cell lung cancer. No further clinical investigation with current dose and schedules is warranted for patients with non-small cell lung cancer who failed first-line platinum-based doublet chemotherapy.


Cancer Research | 2014

Abstract 414: Aberrant DNA methylation and expression of mRNA in EGFR-mutant lung cancer cell line with long-term exposure to gefitinib

Hideki Terai; Kenzo Soejima; Katsuhiko Naoki; Hiroyuki Yasuda; Takashi Sato; Daisuke Arai; Keiko Ohgino; Kota Ishioka; Aoi Kuroda; Tetsuo Tani; Ayano Ohashi; Makoto Nishino; Masayoshi Miyawaki; Junko Hamamoto; Tomoko Betsuyaku

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Many of the patients with non-small cell lung cancer (NSCLC) who initially responded well to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) will eventually relapse. The mechanisms of resistance to EGFR-TKIs is not yet fully clarified. The suppression of genes by DNA methylation is reported to be involved in the mechanism of tolerance to cytotoxic drugs. The purpose of this study is to identify epigenetically regulated genes and to clarify the contribution of epigenetic alteration to the acquired resistance to EGFR-TKI. We established gefitinib-resistant PC-9, which was originally gefitinib-sensitive lung cancer cell line, by serial long term exposure to gefitinib. We collected RNA and DNA from both gefitinib-sensitive and -resistant PC-9 cells and performed comprehensive analysis for DNA methylation and mRNA expression using infinium array and cDNA microarray, respectively. We identified 640 genes those DNA methylations were increased in gefitinib-resistant cell line compared to parental cell line. Then, we selected 29 candidate genes those mRNA expression were decreased in resistant PC-9. We are currently underway to elucidate the specific function of each gene and updated data will be presented at the meeting. Citation Format: Hideki Terai, Kenzo Soejima, Katsuhiko Naoki, Hiroyuki Yasuda, Takashi Sato, Daisuke Arai, Keiko Ohgino, Kota Ishioka, Aoi Kuroda, Tetsuo Tani, Ayano Ohashi, Makoto Nishino, Masayoshi Miyawaki, Junko Hamamoto, Tomoko Betsuyaku. Aberrant DNA methylation and expression of mRNA in EGFR-mutant lung cancer cell line with long-term exposure to gefitinib. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 414. doi:10.1158/1538-7445.AM2014-414


Cancer Research | 2013

Abstract 5652: Activation of FGF2-FGFR1 pathway in EGFR-mutant lung cancer cell line with long-term gefitinib exposure.

Hideki Terai; Kenzo Soejima; Katsuhiko Naoki; Hiroyuki Yasuda; Ryosuke Satomi; Sohei Nakayama; Satoshi Yoda; Shinnosuke Ikemura; Takashi Sato; Kota Ishioka; Daisuke Arai; Keiko Ohgino; Tetsuo Tani; Aoi Kuroda; Junko Hamamoto; Tomoko Betsuyaku

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC Many of the patients with non-small cell lung cancer (NSCLC) with sensitive epidermal growth factor receptor (EGFR)-mutation who initially responded well to EGFR-tyrosine kinase inhibitors (TKIs) eventually relapse. In spite of many studies over the last few years to elucidate this mechanism of acquired resistance to EGFR-TKIs, approximately 30% of the mechanisms of acquired resistance are still unknown. Recently autocrine signaling of fibroblast growth factors (FGFs) and their receptors (FGFRs) has been demonstrated in NSCLC cell lines. And several studies suggest that the FGF-FGFR autocrine growth pathway could be an important mechanism for intrinsic resistance to EGFR-TKIs in NSCLC cell lines with wild-type EGFR. But until now, no report has clarified the role of FGF-FGFR pathway in acquired resistance to EGFR-TKIs in NSCLC cell lines with sensitive EGFR mutations. We have established a gefitinib-resistant cell line (PC9 GR), by serial exposure of gefitinib to PC9, an originally gefitinib-sensitive lung cancer cell line (PC9 na). We confirmed that these cell lines did not harbor two well-known EGFR-TKI resistance mechanisms, the second mutation in the EGFR gene itself, EGFR T790 and the amplification of the MET oncogene. We collected total RNA from both PC9 na and PC9 GR and examined mRNA expression profile, by using cDNA microarray analysis. We found the expressions of FGFR1 and FGF2 were increased in PC9 GR compared to in PC9 na. The growth of PC9 GR cells was inhibited either by PD173074 (inhibitors of FGFRs) or knocking down of FGFR1 or FGF2 by siRNA in combination with gefitinib. FACS analysis revealed that the combination treatment with PD173074 and gefitinib induced apoptosis more efficiently in PC9 GR cells compared to gefitinib alone. PC9 na cells and PC9 GR cells did not show any change in the proportion of apoptotic cells after treatment with PD173074 alone. To further investigate how FGF2-FGFR1 pathway affects resistance to gefitinib in these cell lines, the downstream targets of EGFR signaling including the MEK-ERK and PI3K-AKT pathways were examined. In PC9 na cells, the phosphorylation of EGFR, ERK, and AKT was efficiently inhibited by gefitinib alone. On the other hand, in PC9 GR cells, the phosphorylation of ERK and AKT was not efficiently inhibited by gefitinib alone. However, the inhibition of phosphorylation of ERK was completely and AKT was less efficiently rescued by gefitinib and PD173074 combination therapy. In conclusion, these data suggest the activation of FGF2-FGFR1 signaling pathway contributes to the gefitinib resistance in PC9 GR. FGF2-FGFR1 pathway will be a therapeutic target for a subset of NSCLC that acquires EGFR-TKI resistance. Citation Format: Hideki Terai, Kenzo Soejima, Katsuhiko Naoki, Hiroyuki Yasuda, Ryosuke Satomi, Sohei Nakayama, Satoshi Yoda, Shinnosuke Ikemura, Takashi Sato, Kota Ishioka, Daisuke Arai, Keiko Ohgino, Tetsuo Tani, Aoi Kuroda, Junko Hamamoto, Tomoko Betsuyaku. Activation of FGF2-FGFR1 pathway in EGFR-mutant lung cancer cell line with long-term gefitinib exposure. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 5652. doi:10.1158/1538-7445.AM2013-5652


Archive | 2012

Chemotherapy for Large Cell Neuroendocrine Carcinoma of the Lung: Should It Be Treated with the Same Strategy as Small Cell Lung Carcinoma?

Katsuhiko Naoki; Kenzo Soejima; Takashi Sato; Shinnosuke Ikemura; Hideki Terai; Ryosuke Satomi; Sohei Nakayama; Satoshi Yoda; Koichiro Asano

Lung cancer is leading cause of cancer death in many advanced countries and one of the challenging malignancies because of poor prognosis. Lung cancer is traditionally divided into two major categories, so called small cell lung carcinoma (SCLC) and non-small cell lung carcinoma (NSCLC) because of distinctive prognostic and treatment strategies between them. On the other hand, there is a spectrum of tumors called pulmonary neuroendocrine (NE) tumors that are thought to originate from neuroendocrine cells in the pulmonary and bronchial epithelium. Until recently, pulmonary NE tumors were classified into three categories, i.e., typical carcinoid (TC), atypical carcinoid (AC), and SCLC. Large cell neuroendocrine carcinoma (LCNEC) of the lung was officially identified by Travis et al. in 1991 as a fourth category, a unique higher grade NSCLC existing between TC and SCLC (Travis et al., 1991). It is often difficult to diagnose LCNEC with small biopsy specimens because accurate diagnosis needs morphological and immunohistochemical information. Although earlier reports mainly focused on prognosis after surgical procedures, several recent studies reported on the efficacy of chemotherapy for advanced LCNEC. Because of the limited numbers of cases (in surgical series, LCNEC represents ~3% of lung cancers), large scale prospective studies have not been reported. Standard treatment for LCNEC, especially if advanced, is not established although LCNEC is included in NSCLC in the treatment algorithm in many guidelines. However, accumulating data including recent retrospective studies have suggested that there is similarity in the prognosis and treatment response between LCNEC and SCLC. In this review, we will focus on the treatment of advanced LCNEC for the better selection of chemotherapeutic regimens for the patients with this relatively rare lung cancer.

Collaboration


Dive into the Hideki Terai's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge