Skip Navigation
Skip to contents

Cancer Res Treat : Cancer Research and Treatment

OPEN ACCESS

Author index

Page Path
HOME > Browse articles > Author index
Search
Min Hee Hong 6 Articles
Lung and Thoracic cancer
Aggressive Local Ablative Radiotherapy Mitigates Progression Risk in Oligometastatic Lung Adenocarcinoma
Gowoon Yang, Kyung Hwan Kim, Chang Geol Lee, Min Hee Hong, Hye Ryun Kim, Yeona Cho, Hong In Yoon
Cancer Res Treat. 2024;56(1):115-124.   Published online August 29, 2023
DOI: https://doi.org/10.4143/crt.2023.600
AbstractAbstract PDFSupplementary MaterialPubReaderePub
Purpose
This study aimed to determine the role of local ablative radiotherapy (LART) in oligometastatic/oligoprogressive lung adenocarcinoma.
Materials and Methods
Patients (n=176) with oligometastatic lung adenocarcinoma treated with LART were identified, and those treated with LART at the initial diagnosis of synchronous oligometastatic disease (OMD group) or treated with LART when they presented with repeat oligoprogression (OPD group) were included.
Results
In the OMD group (n=54), the 1- and 3-year progression-free survival (PFS) were 50.9% and 22.5%, respectively, whereas the 1- and 3-year overall survival in the OPD group were 75.9% and 58.1%, respectively. Forty-one patients (75.9%) received LART at all gross disease sites. Tyrosine kinase inhibitor (TKI) use and all-metastatic site LART were significant predictors of higher PFS (p=0.018 and p=0.046, respectively). In patients treated with TKIs at the time of LART (n=23) and those treated with all-metastatic site LART, the 1-year PFS was 86.7%, while that of patients not treated with all-metastatic site LART was 37.5% (p=0.006). In the OPD group (n=122), 67.2% of the patients (n=82) maintained a systemic therapy regimen after LART. The cumulative incidence of changing systemic therapy was 39.6%, 62.9%, and 78.5% at 6 months, 1 year, and 2 years after LART, respectively.
Conclusion
Aggressive LART can be an option to improve survival in patients with oligometastatic disease. Patients with synchronous oligometastatic disease receiving TKI and all-metastatic site LART may have improved PFS. In patients with repeat oligoprogression, LART might potentially extend survival by delaying the need to change the systemic treatment regimen.
  • 2,853 View
  • 181 Download
Close layer
Real World Characteristics and Clinical Outcomes of HER2-Mutant Non–Small Cell Lung Cancer Patients Detected by Next-Generation Sequencing
Beung-Chul Ahn, Ye-Jeong Han, Hye Ryun Kim, Min Hee Hong, Byoung Chul Cho, Sun Min Lim
Cancer Res Treat. 2023;55(2):488-497.   Published online November 9, 2022
DOI: https://doi.org/10.4143/crt.2022.359
AbstractAbstract PDFSupplementary MaterialPubReaderePub
Purpose
This study was conducted to investigate the clinical characteristics of patients with advanced non–small cell lung cancer (NSCLC) harboring human epidermal growth factor receptor 2 (HER2) mutations and to evaluate response to standard treatment and HER2-targeted agents.
Materials and Methods
Using tissue and/or blood next-generation sequencing, we identified 44 patients with NSCLC harboring HER2 mutations who were treated at Severance Hospital between December 2016 and February 2021. Clinical data, including patient characteristics, mutation status, incidence of metastasis for distant lesions, and response to chemotherapy, were retrospectively analyzed.
Results
The median age was 58 years, and 61% of the patients were female. Most patients (64%) were never-smokers. Adenocarcinoma was the most predominant subtype (98%). A total of 66% of the patients had extrathoracic metastatic lesions, and 32% had intracranial lesions at initial presentation. The median time to the development of brain metastasis was 15.6 months (range, 2.4 to 43.7). The most common type of HER2 mutation was 12 base pair in-frame insertion in exon 20, A775_G776insYVMA. Of the 44 patients, two had concomitant driver mutations, one with epidermal growth factor receptor (EGFR) mutation (V769M), and one with BRAF mutation (V600E). Patients treated with pemetrexed-based chemotherapy (75%) had an overall response rate (ORR) and progression-free survival (PFS) of 30% and 8.3 months (95% confidence interval [CI], 3.9 to 12.7), respectively. The ORR and PFS of HER2-targeted agent treated patients (14%) were 0.0% and 1.9 months (95% CI, 0.1 to 2.8), respectively.
Conclusion
Given its distinct characteristics and treatment responses, novel treatment strategies for HER2-mutant NSCLC should be developed promptly to improve survival outcomes of patients.

Citations

Citations to this article as recorded by  
  • Prognostic factors in non-metastatic HER2 ‘low’ and HER2 ‘negative’ breast cancer: single institute experience
    Alper Türkel, Mutlu Doğan, Elif Sertesen, Cengiz Karaçin, Sultan Çiğdem Irkkan, Öztürk Ateş
    Wiener klinische Wochenschrift.2024; 136(11-12): 340.     CrossRef
  • Clinicopathologic and Molecular Characteristics of HER2 (ERBB2)-Altered Non–Small Cell Lung Cancer: Implications for Precision Medicine
    Yurimi Lee, Boram Lee, Yoon-La Choi, Dong-Wook Kang, Joungho Han
    Modern Pathology.2024; 37(6): 100490.     CrossRef
  • Current status and breakthroughs in treating advanced non-small cell lung cancer with EGFR exon 20 insertion mutations
    Meng Hu, Congying Zhong, Jiabing Wang, JinQin Chen, Tao Zhou
    Frontiers in Immunology.2024;[Epub]     CrossRef
  • Efficacy of chemotherapy plus immune checkpoint inhibitors in patients with non-small cell lung cancer who have rare oncogenic driver mutations: a retrospective analysis
    Teppei Yamaguchi, Junichi Shimizu, Reiko Matsuzawa, Naohiro Watanabe, Yoshitsugu Horio, Yutaka Fujiwara
    BMC Cancer.2024;[Epub]     CrossRef
  • Antibody–Drug Conjugates for the Treatment of Non-Small Cell Lung Cancer with Central Nervous System Metastases
    David J. H. Bian, Sara F. Cohen, Anna-Maria Lazaratos, Nathaniel Bouganim, Matthew Dankner
    Current Oncology.2024; 31(10): 6314.     CrossRef
  • Real-World Clinical Outcomes for Patients with EGFR and HER2 Exon 20 Insertion-Mutated Non-Small-Cell Lung Cancer
    Kelly Li, Ian Bosdet, Stephen Yip, Cheryl Ho, Janessa Laskin, Barbara Melosky, Ying Wang, Sophie Sun
    Current Oncology.2023; 30(8): 7099.     CrossRef
  • 5,559 View
  • 268 Download
  • 7 Web of Science
  • 6 Crossref
Close layer
Docetaxel versus Paclitaxel Combined with 5-FU and Leucovorin in Advanced Gastric Cancer: Combined Analysis of Two Phase II Trials
Hong Jae Chon, Sun Young Rha, Chong Kun Im, Chan Kim, Min Hee Hong, Hye Ryun Kim, Jung Ryun An, Sung Hoon Noh, Hyun Cheol Chung, Hei-Cheul Jeung
Cancer Res Treat. 2009;41(4):196-204.   Published online December 31, 2009
DOI: https://doi.org/10.4143/crt.2009.41.4.196
AbstractAbstract PDFPubReaderePub
Purpose

This is an ad hoc analysis of two phase II studies which compared the efficacy and safety of two taxanes (paclitaxel and docetaxel) combined with 5-fluorouracil (5-FU) and leucovorin (LV) in advanced gastric cancer.

Materials and Methods

Patients with advanced gastric adenocarcinoma who were untreated or had only received first-line chemotherapy, were treated with either paclitaxel (PFL; 175 mg/m2) or docetaxel (DFL; 75 mg/m2) on day 1, followed by a bolus of LV (20 mg/m2 days 1~3) and a 24-hour infusion of 5-FU (1,000 mg/m2 days 1~3) every 3 weeks. The primary endpoint was overall response rate (ORR) and the secondary endpoint included survival and toxicity.

Results

Sixty-six patients received DFL (first-line [n=38]; and second-line [n=28]) and 60 patients received PFL (first-line [n=37]; and second-line [n=23]). The ORRs were not significantly different between the 2 groups (DFL, 26%; PFL, 38%). With a median follow-up of 9.5 months, the progression free survival was 5.2 months (95% confidence interval [CI], 4.2~6.5 months) for DFL and 3.3 months (95% CI, 1.3~5.5 months) for PFL (p=0.17). The overall survival was also comparable between the patients who received DFL and PFL (10.0 months [95% CI, 7.2~12.5 months] and 13.9 months [95% CI, 10.9~19.2 months], respectively; p=0.37). The most frequent grade 3~4 adverse event was neutropenia (DFL, 71%; PFL, 62%). DFL and PFL had different non-hematologic toxicities; specifically, grade ≥3 mucositis (5%) and diarrhea (3%) were common in DFL, while nausea/vomiting (15%) and peripheral neuropathy (5%) were common in PFL.

Conclusion

Thus, the two taxanes had similar efficacy in the treatment of advanced gastric cancer, but different toxicity profiles. Prospective comparative studies are required to further clarify the role of taxanes in the treatment of advanced gastric cancer.

Citations

Citations to this article as recorded by  
  • Drug-Induced Peripheral Neuropathy: Diagnosis and Management
    Diala Merheb, Georgette Dib, Maroun Bou Zerdan, Clara El Nakib, Saada Alame, Hazem I. Assi
    Current Cancer Drug Targets.2022; 22(1): 49.     CrossRef
  • Treatment Outcome and Safety of the TCX Regimen for Advanced Gastric Cancer: A Prospective Cohort Study
    Hieu Trong Nguyen, Kien Hung Do, Nguyen Ba Le, Thang Tran
    Cancer Management and Research.2022; Volume 14: 2825.     CrossRef
  • Role for Drug Transporters in Chemotherapy‐Induced Peripheral Neuropathy
    Tore B. Stage, Shuiying Hu, Alex Sparreboom, Deanna L. Kroetz
    Clinical and Translational Science.2021; 14(2): 460.     CrossRef
  • Personalized Antibodies for Gastroesophageal Adenocarcinoma (PANGEA): A Phase II Study Evaluating an Individualized Treatment Strategy for Metastatic Disease
    Daniel V.T. Catenacci, Stephanie Moya, Samantha Lomnicki, Leah M. Chase, Bryan F. Peterson, Natalie Reizine, Lindsay Alpert, Namrata Setia, Shu-Yuan Xiao, John Hart, Uzma D. Siddiqui, D. Kyle Hogarth, Oliver S. Eng, Kiran Turaga, Kevin Roggin, Mitchell C.
    Cancer Discovery.2021; 11(2): 308.     CrossRef
  • Neuroinflammatory Process Involved in Different Preclinical Models of Chemotherapy-Induced Peripheral Neuropathy
    Giulia Fumagalli, Laura Monza, Guido Cavaletti, Roberta Rigolio, Cristina Meregalli
    Frontiers in Immunology.2021;[Epub]     CrossRef
  • Pathogenesis of paclitaxel-induced peripheral neuropathy: A current review of in vitro and in vivo findings using rodent and human model systems
    Nathan P. Staff, Jill C. Fehrenbacher, Martial Caillaud, M. Imad Damaj, Rosalind A. Segal, Sandra Rieger
    Experimental Neurology.2020; 324: 113121.     CrossRef
  • Salvage systemic therapy for advanced gastric and oesophago-gastric junction adenocarcinoma
    Yoko Tomita, Max Moldovan, Rachael Chang Lee, Amy HC Hsieh, Amanda Townsend, Timothy Price
    Cochrane Database of Systematic Reviews.2020;[Epub]     CrossRef
  • Paclitaxel, 5-fluorouracil, and leucovorin combination chemotherapy as first-line treatment in patients with advanced gastric cancer
    Wan-Cai Que, Yan-Fang Huang, Xiao-Yan Lin, Yan-Qin Lan, Xin-Yan Gao, Xin-Li Wang, Ri-Ping Wu, Bin Du, Xiao-Bin Huang, Hong-qiang Qiu, Dong-Ta Zhong
    Anti-Cancer Drugs.2019; 30(3): 302.     CrossRef
  • Randomised phase II trial comparing four front-line doublets in Asian patients with metastatic gastric cancer
    Chan Kim, Hong Jae Chon, Joo Hoon Kim, Minkyu Jung, Chung Mo Nam, Hyo Song Kim, Beodeul Kang, Hyun Cheol Chung, Sun Young Rha
    European Journal of Cancer.2019; 112: 20.     CrossRef
  • Trastuzumab emtansine versus taxane use for previously treated HER2-positive locally advanced or metastatic gastric or gastro-oesophageal junction adenocarcinoma (GATSBY): an international randomised, open-label, adaptive, phase 2/3 study
    Peter C Thuss-Patience, Manish A Shah, Atsushi Ohtsu, Eric Van Cutsem, Jaffer A Ajani, Hugo Castro, Wasat Mansoor, Hyun Cheol Chung, Gyorgy Bodoky, Kohei Shitara, Gail D Lewis Phillips, Tina van der Horst, Marie-Laurence Harle-Yge, Betsy L Althaus, Yoon-K
    The Lancet Oncology.2017; 18(5): 640.     CrossRef
  • Pathophysiology of Chemotherapy-Induced Peripheral Neuropathy
    Hana Starobova, Irina Vetter
    Frontiers in Molecular Neuroscience.2017;[Epub]     CrossRef
  • Identifying therapeutic targets in gastric cancer: the current status and future direction
    Beiqin Yu, Jingwu Xie
    Acta Biochimica et Biophysica Sinica.2016; 48(1): 90.     CrossRef
  • Role of Transient Receptor Potential Channels in Paclitaxel- and Oxaliplatin-induced Peripheral Neuropathy
    Kyoji Taguchi
    YAKUGAKU ZASSHI.2016; 136(2): 287.     CrossRef
  • Paclitaxel combined with oxaliplatin as first-line chemotherapy for locally advanced or metastatic gastric cancer
    Chunmei Shi, Qiang Chen, Songfei Shen, Riping Wu, Baoyu Yang, Qing Liu, Qian Xu
    Expert Review of Anticancer Therapy.2015; 15(5): 595.     CrossRef
  • Corosolic acid enhances 5-fluorouracil-induced apoptosis against SNU-620 human gastric carcinoma cells by inhibition of mammalian target of rapamycin
    HYUN SU LEE, JUN BEOM PARK, MYUNG SUN LEE, EUN YOUNG CHA, JI YEON KIM, JI YOUNG SUL
    Molecular Medicine Reports.2015; 12(3): 4782.     CrossRef
  • Paclitaxel-based regimens as first-line treatment in advanced gastric cancer
    Zengqing Guo, Xiaojie Wang, Rongbo Lin, Ling Chen, Nanfeng Fan, Yu Chen, Jinyuan Lin, Jiami Yu
    Journal of Chemotherapy.2015; 27(2): 94.     CrossRef
  • A phase 2 study of fluorouracil/leucovorin in combination with paclitaxel and oxaliplatin as a salvage treatment in patients with refractory or relapsed advanced gastric cancer
    Rongbo Lin, Nanfeng Fan, Guangfeng Wu, Ying Chen, Zengqing Guo, Xiaojie Wang, Feng Jin, Ling Chen, Jie Liu
    Journal of Chemotherapy.2015; 27(1): 52.     CrossRef
  • Paclitaxel combined with capecitabine as first-line chemotherapy for advanced or recurrent gastric cancer
    MEIQIN YUAN, YUNSHAN YANG, WANGXIA LV, ZHENGBO SONG, HAIJUN ZHONG
    Oncology Letters.2014; 8(1): 351.     CrossRef
  • Chemotherapy-induced peripheral neurotoxicity (CIPN): An update
    Andreas A. Argyriou, Jordi Bruna, Paola Marmiroli, Guido Cavaletti
    Critical Reviews in Oncology/Hematology.2012; 82(1): 51.     CrossRef
  • Peripheral neuropathies from chemotherapeutics and targeted agents: diagnosis, treatment, and prevention
    W. Grisold, G. Cavaletti, A. J. Windebank
    Neuro-Oncology.2012; 14(suppl 4): iv45.     CrossRef
  • Successful Treatment of a Patient with HER2-Positive Metastatic Gastric Cancer with Third-Line Combination Therapy with Irinotecan, 5-Fluorouracil, Leucovorin and Trastuzumab (FOLFIRI-T)
    Florian Weissinger, Marc Reymond, Klaus Dumke, Martin Krüger
    Onkologie.2011; 34(10): 548.     CrossRef
  • RAD001 shows activity against gastric cancer cells and overcomes 5-FU resistance by downregulating thymidylate synthase
    Kyung-Hun Lee, Hyung-Seok Hur, Seock-Ah Im, Juhee Lee, Hwang-Phill Kim, Young-Kwang Yoon, Sae-Won Han, Sang-Hyun Song, Do-Youn Oh, Tae-You Kim, Yung-Jue Bang
    Cancer Letters.2010; 299(1): 22.     CrossRef
  • 12,083 View
  • 76 Download
  • 22 Crossref
Close layer
Clinical Value of Ezrin Expression in Primary Osteosarcoma
Chan Kim, Eunah Shin, Soojung Hong, Hong Jae Chon, Hye Ryun Kim, Jung Ryun Ahn, Min Hee Hong, Woo Ick Yang, Jae Kyung Roh, Sun Young Rha
Cancer Res Treat. 2009;41(3):138-144.   Published online September 28, 2009
DOI: https://doi.org/10.4143/crt.2009.41.3.138
AbstractAbstract PDFPubReaderePub
Purpose

Ezrin is a membrane cytoskeletal linker protein and it is known to be associated with metastasis of primary osteosarcoma. The aim of this study is to determine the relationship between an ezrin expression and several key clinical parameters and to elucidate its potential prognostic value for patients with osteosarcoma.

Materials and Methods

Seventy patients with histologically confirmed osteosarcoma and who had no distant metastasis were enrolled between 1995 and 2005 at Yonsei Cancer Center, Severance Hospital, Korea. The clinical parameters were retrospectively reviewed and immunohistochemical staining (IHC) for ezrin was performed using the surgically resected specimens.

Results

Of the 70 tumor specimens, 39 (55.7%) revealed an ezrin expression. More of an osteoblastic histology and an elevated initial ALP level were observed in the ezrin positive patients than in the ezrin negative patients (p=0.008 and 0.001, respectively). The proportion of patients who favorably responded to neoadjuvant chemotherapy (≥90% necrosis) was significantly higher in the group of ezrin positive patients than that in the group of ezrin negative patient (72.2% vs 45.2%, respectively, p=0.024). The ezrin positive patients showed more frequent recurrence than did the ezrin negative patients (64.1% vs 35.5%, respectively, p=0.017). The patients with an ezrin expression also demonstrated poorer survival than did those patients without ezrin expression (5-year EFS: 31.7% vs 61.3%, respectively, p=0.023, 5-year OS: 53.4% vs 71.0%, respectively, p=0.022). When comparing EFS according to both an ezrin expression and chemoresponsiveness, there were trends that the ezrin negative/chemoresponsive group showed the best 5-year EFS (71.4%), followed by the ezrin negative/chemoresistant group (52.9%), the ezrin positive/chemoresponsive group (38.1%) and the ezrin positive/chemoresistant group (13.6%). These trends were statistically significant (p=0.036).

Conclusion

The expression of ezrin by IHC staining was found in 55.7% of the patients with metastasis-free osteosarcoma. Immunoreactivity to ezrin is a negative prognostic factor for survival for the patients suffering with osteosarcoma. Identifying an ezrin expression might offer a valuable piece of information when treating patients with primary osteosarcoma.

Citations

Citations to this article as recorded by  
  • Osteosarcoma-Specific Genes as a Diagnostic Tool and Clinical Predictor of Tumor Progression
    Pattaralawan Sittiju, Parunya Chaiyawat, Dumnoensun Pruksakorn, Jeerawan Klangjorhor, Weerinrada Wongrin, Phichayut Phinyo, Rawikant Kamolphiwong, Areerak Phanphaisarn, Pimpisa Teeyakasem, Prachya Kongtawelert, Peraphan Pothacharoen
    Biology.2022; 11(5): 698.     CrossRef
  • Ezrin gone rogue in cancer progression and metastasis: An enticing therapeutic target
    Ganesh Kumar Barik, Osheen Sahay, Debasish Paul, Manas Kumar Santra
    Biochimica et Biophysica Acta (BBA) - Reviews on Cancer.2022; 1877(4): 188753.     CrossRef
  • Mass Spectrometric-Based Proteomics for Biomarker Discovery in Osteosarcoma: Current Status and Future Direction
    Nutnicha Sirikaew, Dumnoensun Pruksakorn, Parunya Chaiyawat, Somchai Chutipongtanate
    International Journal of Molecular Sciences.2022; 23(17): 9741.     CrossRef
  • Ezrin is a prognostic biomarker in patients with clear cell metastatic renal cell carcinoma receiving sunitinib
    Bulent Cetin, Ipek Isik Gonul, Ozge Gumusay, Baris Afsar, Irem Bilgetekin, Ahmet Ozet, Aytug Uner
    Journal of Cancer Research and Therapeutics.2021; 17(2): 408.     CrossRef
  • High expression of EZR (ezrin) gene is correlated with the poor overall survival of breast cancer patients
    Rongju Zhang, Shaohui Zhang, Rongge Xing, Qin Zhang
    Thoracic Cancer.2019; 10(10): 1953.     CrossRef
  • The clinical prognostic significance of ezrin in patients with bone and soft tissue sarcomas: a meta‐analysis
    Feng Wang, Tao Yu, Chengbin Ma, Haifei Zhang, Zhiyu Zhang
    FEBS Open Bio.2019; 9(10): 1744.     CrossRef
  • Re-calculating! Navigating through the osteosarcoma treatment roadblock
    J. McGuire, T.J. Utset-Ward, D.R. Reed, C.C. Lynch
    Pharmacological Research.2017; 117: 54.     CrossRef
  • Correlation of Ezrin Expression Pattern and Clinical Outcomes in Ewing Sarcoma
    Thomas Cash, Hong Yin, Courtney McCracken, Zhi Geng, Steven G. DuBois, Bahig M. Shehata, Thomas A. Olson, Howard M. Katzenstein, Cynthia Wetmore
    Sarcoma.2017; 2017: 1.     CrossRef
  • Differentially activated Src kinase in chemo‐naïve human primary osteosarcoma cells and effects of a Src kinase inhibitor
    Marcella Laschi, Giulia Bernardini, Michela Geminiani, Fabrizio Manetti, Mattia Mori, Adriano Spreafico, Domenico Campanacci, Rodolfo Capanna, Silvia Schenone, Maurizio Botta, Annalisa Santucci
    BioFactors.2017; 43(6): 801.     CrossRef
  • The Prognostic Role of Ezrin and HER2/neu Expression in Osteosarcoma
    Asmaa G. Abdou, Mona Kandil, Nancy Y. Asaad, Marwa M. Dawoud, Ahmed A. Shahin, Amal F. Abd Eldayem
    Applied Immunohistochemistry & Molecular Morphology.2016; 24(5): 355.     CrossRef
  • Establishment of Four New Human Primary Cell Cultures from Chemo‐Naïve Italian Osteosarcoma Patients
    Marcella Laschi, Giulia Bernardini, Michela Geminiani, Lorenzo Ghezzi, Loredana Amato, Daniela Braconi, Lia Millucci, Bruno Frediani, Adriano Spreafico, Alessandro Franchi, Domenico Campanacci, Rodolfo Capanna, Annalisa Santucci
    Journal of Cellular Physiology.2015; 230(11): 2718.     CrossRef
  • Prognostic Significance of Serum Alkaline Phosphatase Level in Osteosarcoma: A Meta-Analysis of Published Data
    Hai-Yong Ren, Ling-Ling Sun, Heng-Yuan Li, Zhao-Ming Ye
    BioMed Research International.2015; 2015: 1.     CrossRef
  • Prognostic Value of Ezrin in Various Cancers: A Systematic Review and Updated Meta-analysis
    Jianwei Li, Kuanhai Wei, Hailang Yu, Dan Jin, Gang Wang, Bin Yu
    Scientific Reports.2015;[Epub]     CrossRef
  • Alteration of WWOX in human cancer, a clinical view
    Izabela Baryła, Ewa Styczeń-Binkowska, Andrzej K Bednarek
    Experimental Biology and Medicine.2015; 240(3): 305.     CrossRef
  • The prognostic value of elevated ezrin in patients with osteosarcoma
    Deng-Xing Lun, Yong-Cheng Hu, Zhao-Wan Xu, Li-Na Xu, Bin-Wu Wang
    Tumor Biology.2014; 35(2): 1263.     CrossRef
  • Prognostic role of cytovillin expression in patients with osteosarcoma: a meta-analysis
    Shibing Guo, Rui Bai, Wei Zhao, Yuxin Wang, Zhenqun Zhao, Wei Feng
    Tumor Biology.2014; 35(1): 469.     CrossRef
  • CD99 suppresses osteosarcoma cell migration through inhibition of ROCK2 activity
    C Zucchini, M C Manara, R S Pinca, P De Sanctis, C Guerzoni, M Sciandra, P-L Lollini, G Cenacchi, P Picci, L Valvassori, K Scotlandi
    Oncogene.2014; 33(15): 1912.     CrossRef
  • Correlations of Ezrin Expression with Pathological Characteristics and Prognosis of Osteosarcoma: A Meta-Analysis
    Da-Hang Zhao, Jiang Zhu, Wen-Bo Wang, Feng Dong, Qiao Zhang, Hong-Wu Fan, Jing-Zhe Zhang, Yong-Ming Wang
    The Scientific World Journal.2014; 2014: 1.     CrossRef
  • Expression of HMGA1 and Ezrin in laryngeal squamous cell carcinoma
    De-Sheng Wang, Chun-Chen Pan, Hai-Chun Lai, Jian-Min Huang
    Acta Oto-Laryngologica.2013; 133(6): 626.     CrossRef
  • The β5/focal adhesion kinase/glycogen synthase kinase 3β integrin pathway in high-grade osteosarcoma: a protein expression profile predictive of response to neoadjuvant chemotherapy
    Sophie Le Guellec, Elizabeth Cohen-Jonathan Moyal, Thomas Filleron, Marie-Bernadette Delisle, Christine Chevreau, Hervé Rubie, Marie-Pierre Castex, Jerome Sales de Gauzy, Paul Bonnevialle, Anne Gomez-Brouchet
    Human Pathology.2013; 44(10): 2149.     CrossRef
  • Meta-analysis of Associations of the Ezrin Gene with Human Osteosarcoma Response to Chemotherapy and Prognosis
    Zhe Wang, Mao-Lin He, Jin-Min Zhao, Hai-Hui Qing, Yang Wu
    Asian Pacific Journal of Cancer Prevention.2013; 14(5): 2753.     CrossRef
  • The Prognostic Role of Ezrin Immunoexpression in Osteosarcoma: A Meta-Analysis of Published Data
    Hongtao Li, Daliu Min, Hui Zhao, Zhiyu Wang, Weixiang Qi, Shuier Zheng, Lina Tang, Aina He, Yuanjue Sun, Yang Yao, Zan Shen, David Loeb
    PLoS ONE.2013; 8(6): e64513.     CrossRef
  • Effects of siRNA-mediated Cdc2 silencing on MG63 cell proliferation and apoptosis
    XIANG-YONG QUE, YI LI, YU HAN, XIN-ZHI LI
    Molecular Medicine Reports.2013; 7(2): 466.     CrossRef
  • Clinicopathologic Implication of Ezrin Expression in Non-small Cell Lung Cancer
    Ho Won Lee, Eui Han Kim, Mee-Hye Oh
    Korean Journal of Pathology.2012; 46(5): 470.     CrossRef
  • Effects of transplantation sites on tumour growth, pulmonary metastasis and ezrin expression of canine osteosarcoma cell lines in nude mice
    T. Jaroensong, Y. Endo, S.‐J. Lee, A. Kamida, M. Mochizuki, R. Nishimura, N. Sasaki, T. Nakagawa
    Veterinary and Comparative Oncology.2012; 10(4): 274.     CrossRef
  • Prognostic implication of ezrin expression in esophageal squamous cell carcinoma
    Jian‐Jun Xie, Li‐Yan Xu, Zhi‐Yong Wu, Qing Zhao, Xiu‐E Xu, Jian‐Yi Wu, Qiao Huang, En‐Min Li
    Journal of Surgical Oncology.2011; 104(5): 538.     CrossRef
  • Identification of the VIL2 enhancer in human embryonic kidney cells
    Shu‐Ying Gao, Yan‐Peng Dai, Xia Long, Fang‐Ting Zhang, Jie Yu
    Cell Biology International.2011; 35(10): 967.     CrossRef
  • Rôle du pathologiste dans la prise en charge des tumeurs osseuses primitives malignes : ostéosarcomes et tumeurs de la famille Ewing après traitement néoadjuvant
    Anne Gomez-Brouchet, Corinne Bouvier, Anne-Valérie Decouvelaere, Frédérique Larousserie, Sébastien Aubert, Xavier Leroy, Jean-Marc Guinebretière, Aurore Coulomb, Elisabeth Cassagnau, Anne de Muret, Virginie Audard, Béatrice Marie, Gonzague de Pinieux
    Annales de Pathologie.2011; 31(6): 455.     CrossRef
  • Potential transcriptional regulatory regions exist upstream of the human ezrin gene promoter in esophageal carcinoma cells
    S. Gao, Y. Dai, M. Yin, J. Ye, G. Li, J. Yu
    Acta Biochimica et Biophysica Sinica.2011; 43(6): 455.     CrossRef
  • YY1 overexpression is associated with poor prognosis and metastasis-free survival in patients suffering osteosarcoma
    Filomena de Nigris, Licciana Zanella, Francesco Cacciatore, Anna De Chiara, Flavio Fazioli, Gennaro Chiappetta, Gaetano Apice, Teresa Infante, Mario Monaco, Raffaele Rossiello, Gaetano De Rosa, Marco Alberghini, Claudio Napoli
    BMC Cancer.2011;[Epub]     CrossRef
  • 17β-Estradiol Enhances Breast Cancer Cell Motility and Invasion via Extra-Nuclear Activation of Actin-Binding Protein Ezrin
    Shuhui Zheng, Jinghe Huang, Kewen Zhou, Chengxi Zhang, Qiuling Xiang, Zhi Tan, Tinghuai Wang, Xiaodong Fu, Jean-Marc Vanacker
    PLoS ONE.2011; 6(7): e22439.     CrossRef
  • Ezrin expression predicts local recurrence and development of metastases in soft tissue sarcomas
    Ana Carneiro, Pär-Ola Bendahl, Måns Åkerman, Henryk A Domanski, Anders Rydholm, Jacob Engellau, Mef Nilbert
    Journal of Clinical Pathology.2011; 64(8): 689.     CrossRef
  • Immunoexpression of Ezrin and CD44 in Patients With Osteosarcoma
    Erica Boldrini, Stela Verzinhasse Peres, Sandra Morini, Beatriz de Camargo
    Journal of Pediatric Hematology/Oncology.2010; 32(6): e213.     CrossRef
  • Prognostic and predictive biomarkers of canine osteosarcoma
    Gayathri Thevi Selvarajah, Jolle Kirpensteijn
    The Veterinary Journal.2010; 185(1): 28.     CrossRef
  • 11,137 View
  • 78 Download
  • 34 Crossref
Close layer
Prognosis of pN3 Stage Gastric Cancer
Jung Ryun Ahn, Minkyu Jung, Chan Kim, Min Hee Hong, Hong Jae Chon, Hye Ryun Kim, Hei-Cheul Jeung, Woo Jin Hyung, Sung Sook Lee, Hyun Cheol Chung, Sung Hoon Noh, Sun Young Rha
Cancer Res Treat. 2009;41(2):73-79.   Published online June 30, 2009
DOI: https://doi.org/10.4143/crt.2009.41.2.73
AbstractAbstract PDFPubReaderePub
Purpose

The aim of this study was to determine the prognosis of pN3 stage gastric cancer patients after they have undergone curative resection, and we also wanted to identify the prognostic factors according to the clinico-pathologic features.

Materials and Methods

Between January 2000 and December 2004, we retrospectively reviewed the medical records of the patients with histologically confirmed pN3 stage gastric cancer. They underwent both gastrectomy and lymphadenectomy with a curative aim. We categorized the pN3 stage patients into 2 groups; one with pN3 only (pN3M0) and the other with pN3 combined with M1 stage (pN3M1) that included peritoneal seeding, hepatic metastasis or para-aortic LN metastasis.

Results

Out of 467 patients with stage IV gastric adenocarcinoma who received surgery, 260 patients underwent curative resection and they were pathologically staged as N3. Among these 260 patients, 78 patients were classified as the pN3/M1 stage. For all the patients, the median follow-up period was 19 months (range: 1~108 months) and the median overall survival time was 16.2 months (95% CI, 14.1~18.3%). The 5-year survival rate of the pN3/M0 group was significantly higher than that of the pN3/M1 group (12.6% vs. 2.6%, respectively, p<0.0001). The identified predictor for a worse prognosis was an advanced T4 stage (HR: 3.38, 95% CI, 1.4~8.3, p=0.008) for the pN3 patients.

Conclusion

The survival for the pN3 gastric cancer patients after curative gastrectomy was significantly longer in the pN3/M0 group as compared to that of the pN3/M1 group. An advanced T stage was a predictor for a poor prognosis for the pN3 patients. Therefore, diverse treatment strategies for these heterogeneous pN3 gastric cancer patients are needed for improving their survival.

Citations

Citations to this article as recorded by  
  • GSK461364A suppresses proliferation of gastric cancer cells and induces apoptosis
    Dilara Ataseven, Şeyma Taştemur, Fatih Yulak, Sebahattin Karabulut, Mustafa Ergul
    Toxicology in Vitro.2023; 90: 105610.     CrossRef
  • HDAC4 promotes the growth and metastasis of gastric cancer via autophagic degradation of MEKK3
    Wei-Jie Zang, Yi-Lin Hu, Chen-Yu Qian, Ying Feng, Jia-Zhou Liu, Jun-Ling Yang, Hua Huang, Yi-Zhun Zhu, Wan-Jiang Xue
    British Journal of Cancer.2022; 127(2): 237.     CrossRef
  • Tumor purity as a prognosis and immunotherapy relevant feature in gastric cancer
    Zhe Gong, Jieyun Zhang, Weijian Guo
    Cancer Medicine.2020; 9(23): 9052.     CrossRef
  • S100A10 Accelerates Aerobic Glycolysis and Malignant Growth by Activating mTOR-Signaling Pathway in Gastric Cancer
    Yan Li, Xiao-Yu Li, Li-Xiang Li, Ru-Chen Zhou, Yinhe Sikong, Xiang Gu, Bi-Ying Jin, Bing Li, Yan-Qing Li, Xiu-Li Zuo
    Frontiers in Cell and Developmental Biology.2020;[Epub]     CrossRef
  • Oncología personalizada: principales biomarcadores en el pronóstico y tratamiento de tumores sólidos
    Marta Molina Romero, Emilio José Laserna Mendieta, Gema María Varo Sánchez, María Concepción Alonso-Cerezo, María Orera Clemente
    Revista del Laboratorio Clínico.2019; 12(3): e1.     CrossRef
  • Is it worthy of adding dissection of the superior mesenteric vein lymph node (14v) to standard D2 gastrectomy for distal gastric cancers with No. 6 lymph node metastasis?
    J. Zhang, S. Zou, R. Luo, Z. Zhu, Z. Wang, H. Xu, B. Huang
    Clinical and Translational Oncology.2019; 21(12): 1699.     CrossRef
  • Reduced expression of Rap1GAP as a prognostic biomarker for primary gastric cancer patients
    Jingjing Zhao, Cong Mai, Desheng Weng, Changlong Chen, Ziqi Zhou, Yuan Liu, Zhiwei Zhou, Peng Wang
    Cancer Biomarkers.2018; 22(3): 375.     CrossRef
  • Prognostic Discrepancy of the 6th and 7th UICC N Classification for Lymph Node Staging in Gastric Cancer Patients after Curative Resection
    Sung Jin Oh, Byoung Jo Suh, Jong Kwon Park, Sung Don Oh, Hang Jong Yu
    Case Reports in Oncology.2017; 10(1): 57.     CrossRef
  • Gastric biomarkers: a global review
    Nick Baniak, Jenna-Lynn Senger, Shahid Ahmed, S. C. Kanthan, Rani Kanthan
    World Journal of Surgical Oncology.2016;[Epub]     CrossRef
  • MiR 21‐5p as a predictor of recurrence in young gastric cancer patients
    Soo‐Kyung Park, Young Soo Park, Ji Yong Ahn, Eun‐Ju Do, Dongho Kim, Jee Eun Kim, kyoungwon Jung, Jeong‐Sik Byeon, Byong Duk Ye, Dong Hoon Yang, Sang Hyoung Park, Sung Wook Hwang, Hwoon‐Yong Jung, Seung‐Jae Myung
    Journal of Gastroenterology and Hepatology.2016; 31(8): 1429.     CrossRef
  • Functional polymorphisms in apoptosis pathway genes and survival in patients with gastric cancer
    Dongying Gu, Mulong Du, Cuiju Tang, Haiyan Chu, Zhi Xu, Xinyin Huo, Weida Gong, Yongfei Tang, Jianwei Zhou, Na Tong, Yong Xu, Zhengdong Zhang, Meilin Wang, Jinfei Chen
    Environmental and Molecular Mutagenesis.2014; 55(5): 421.     CrossRef
  • Improved survival after adding dissection of the superior mesenteric vein lymph node (14v) to standard D2 gastrectomy for advanced distal gastric cancer
    Bang Wool Eom, Jungnam Joo, Young-Woo Kim, Daniel Reim, Ji Yeon Park, Hong Man Yoon, Keun Won Ryu, Jong Yeul Lee, Myeong-Cherl Kook
    Surgery.2014; 155(3): 408.     CrossRef
  • Comprehensive genomic meta-analysis identifies intra-tumoural stroma as a predictor of survival in patients with gastric cancer
    Yonghui Wu, Heike Grabsch, Tatiana Ivanova, Iain Beehuat Tan, Jacinta Murray, Chia Huey Ooi, Alexander Ian Wright, Nicholas P West, Gordon G A Hutchins, Jeanie Wu, Minghui Lee, Julian Lee, Jun Hao Koo, Khay Guan Yeoh, Nicole van Grieken, Bauke Ylstra, Sun
    Gut.2013; 62(8): 1100.     CrossRef
  • Validity and Necessity of Sub-classification of N3 in the 7thUICC TNM Stage of Gastric Cancer
    Fang-Xuan Li, Ru-Peng Zhang, Han Liang, Ji-Chuan Quan, Hui Liu, Hui Zhang
    Asian Pacific Journal of Cancer Prevention.2013; 14(3): 2091.     CrossRef
  • Is the New Seventh AJCC/UICC Staging System Appropriate for Patients with Gastric Cancer?
    Hong Man Yoon, Keun Won Ryu, Byung Ho Nam, Soo Jeong Cho, Sook Ryun Park, Jong Yeul Lee, Jun Ho Lee, Myeong-Cherl Kook, Il Ju Choi, Young-Woo Kim
    Journal of the American College of Surgeons.2012; 214(1): 88.     CrossRef
  • Comparison of the 6th and 7th Editions of the UICC TNM Staging System for Gastric Cancer: Results of a Chinese Single-Institution Study of 1,503 Patients
    Wei Wang, Xiao-wei Sun, Chao-feng Li, Lin Lv, Yuan-fang Li, Ying-bo Chen, Da-zhi Xu, Rajiv Kesari, Chun-yu Huang, Wei Li, You-qing Zhan, Zhi-wei Zhou
    Annals of Surgical Oncology.2011; 18(4): 1060.     CrossRef
  • 11,870 View
  • 76 Download
  • 16 Crossref
Close layer
Treatment Outcomes of Sunitinib Treatment in Advanced Renal Cell Carcinoma Patients: A Single Cancer Center Experience in Korea
Min Hee Hong, Hyo Song Kim, Chan Kim, Jung Ryun Ahn, Hong Jae Chon, Sang-Joon Shin, Joong-Bae Ahn, Hyun Cheol Chung, Sun Young Rha
Cancer Res Treat. 2009;41(2):67-72.   Published online June 30, 2009
DOI: https://doi.org/10.4143/crt.2009.41.2.67
AbstractAbstract PDFPubReaderePub
Purpose

The retrospective study was performed to assess the efficacy and toxicity profiles of sunitinib in Korean patients with metastatic renal cell carcinoma (RCC).

Materials and Methods

Between January 2005 and December 2008, 76 Korean patients with recurrent/metastatic RCC who received sunitinib were retrospectively reviewed. The primary end point was progression-free survival and the secondary end points were overall survival and response rate. We also assessed the toxicities associated with sunitinib treatment.

Results

Of the 76 patients, 69 (90.1%) were diagnosed with clear cell RCC. The median progression-free survival and overall survival were 7.2 and 22.8 months, respectively in overall patients. Sixty-two patients (81.6%) received 50 mg 4 week and 2 week off schedule, and 14 patients (18.4%) received 37.5 mg daily on a daily continuous schedule. The objective response rate and disease control rate were 27.6% and 84.2%, respectively. A dose reduction or reduction in dose due to adverse events occurred in 76% of the patients, whereas 11% of the patients had discontinued treatment. Other common laboratory abnormalities were increased serum creatinine (75.6%), elevated alanine aminotransferase (71.0%), neutropenia (61.8%), anemia (69.7%), and increased aspartate aminotrasferase (53.3%). Grade 3/4 toxicities occurred as follows: thrombocytopenia (38.2%), fatigue (10.5%), stomatitis (10.5%), and hand-foot syndrome (9.2%).

Conclusion

Our results indicate that sunitinib treatment is effective and tolerable for ecurrent/metastatic RCC patients in Korea. Further studies with prognostic or biochemical factors are needed to clarify the different toxicity profiles of this study.

Citations

Citations to this article as recorded by  
  • pH-Responsive Block Copolymer Micelles of Temsirolimus: Preparation, Characterization and Antitumor Activity Evaluation
    Ling Wang, Fangqing Cai, Yixuan Li, Xiaolan Lin, Yuting Wang, Weijie Liang, Caiyu Liu, Cunze Wang, Junshan Ruan
    International Journal of Nanomedicine.2024; Volume 19: 9821.     CrossRef
  • Comparative Evaluation of Safety and Efficacy of Alternate Schedule (AS) of Sunitinib in Asian and Non-Asian Patient Population for the Treatment of Metastatic Renal Cell Cancer (mRCC): A Meta-Analysis
    Amit Joshi, Ishan Patel, Pratiksha Kapse, Manmohan Singh
    Kidney Cancer.2022; 6(1): 37.     CrossRef
  • High Serum Levels of IL-6 Predict Poor Responses in Patients Treated with Pembrolizumab plus Axitinib for Advanced Renal Cell Carcinoma
    Yun Beom Sang, Hannah Yang, Won Suk Lee, Seung Joon Lee, Seul-Gi Kim, Jaekyung Cheon, Beodeul Kang, Chang Woo Kim, Hong Jae Chon, Chan Kim
    Cancers.2022; 14(23): 5985.     CrossRef
  • Prognostic factors for overall survival in patients with clear cell metastatic renal cell carcinoma
    Dongrul Shin, Chang Wook Jeong, Cheryn Song, Minyong Kang, Seong Il Seo, Jung Kwon Kim, Hakmin Lee, Jinsoo Chung, Sung-Hoo Hong, Eu Chang Hwang, Cheol Kwak, Jae Young Park
    Medicine.2021; 100(31): e26826.     CrossRef
  • Efficacy and Prognostic Factors of Sunitinib as First-Line Therapy for Patients With Metastatic Renal Cell Carcinoma in an Arab Population
    Ahmed Badran, Mahmoud A. Elshenawy, Amgad Shahin, Ali Aljubran, Ahmed Alzahrani, Abdelmoneim Eldali, Shouki Bazarbashi
    JCO Global Oncology.2020; (6): 19.     CrossRef
  • Avelumab plus axitinib vs sunitinib for advanced renal cell carcinoma: Japanese subgroup analysis from JAVELIN Renal 101
    Motohide Uemura, Yoshihiko Tomita, Hideaki Miyake, Shingo Hatakeyama, Hiro‐omi Kanayama, Kazuyuki Numakura, Toshio Takagi, Tomoyuki Kato, Masatoshi Eto, Wataru Obara, Hirotsugu Uemura, Toni K. Choueiri, Robert J. Motzer, Yosuke Fujii, Yoichi Kamei, Yoshik
    Cancer Science.2020; 111(3): 907.     CrossRef
  • First-Line Axitinib Versus Sorafenib in Asian Patients with Metastatic Renal Cell Carcinoma: Exploratory Subgroup Analyses of Phase III Data
    Xinan Sheng, Feng Bi, Xiubao Ren, Ying Cheng, Jinwan Wang, Brad Rosbrook, Ming Jiang, Jun Guo
    Future Oncology.2019; 15(1): 53.     CrossRef
  • The Roles of Common Variation and Somatic Mutation in Cancer Pharmacogenomics
    Hiu Ting Chan, Yoon Ming Chin, Siew-Kee Low
    Oncology and Therapy.2019; 7(1): 1.     CrossRef
  • Risk of fatal adverse events in cancer patients treated with sunitinib
    Bin Zhao, Hong Zhao, Jiaxin Zhao
    Critical Reviews in Oncology/Hematology.2019; 137: 115.     CrossRef
  • Temsirolimus in Asian Metastatic/Recurrent Non-clear Cell Renal Carcinoma
    Jii Bum Lee, Hyung Soon Park, Sejung Park, Hyo Jin Lee, Kyung A Kwon, Young Jin Choi, Yu Jung Kim, Chung Mo Nam, Nam Hoon Cho, Beodeul Kang, Hyun Cheol Chung, Sun Young Rha
    Cancer Research and Treatment.2019; 51(4): 1578.     CrossRef
  • Stomatitis and VEGFR-Tyrosine Kinase Inhibitors (VR-TKIs): A Review of Current Literature in 4369 Patients
    Claudia Arena, Giuseppe Troiano, Alfredo De Lillo, Nunzio F. Testa, Lorenzo Lo Muzio
    BioMed Research International.2018; 2018: 1.     CrossRef
  • Efficacy, safety, and prognostic indicators of first-line sunitinib in patients with metastatic renal cell carcinoma
    Nahjatul Kursyiah Abd Ghafar, Adlinda Alip, Teng Aik Ong, Ning Yi Yap, Marniza Saad
    Journal of Cancer Research and Therapeutics.2018; 14(6): 1303.     CrossRef
  • Assessment of ethnic differences in sunitinib outcome between Caucasian and Asian patients with metastatic renal cell carcinoma: a meta-analysis
    Xiaoyan Liu, Marta Fiocco, Jesse J. Swen, Henk-Jan Guchelaar
    Acta Oncologica.2017; 56(4): 582.     CrossRef
  • Oncogenic miR-100-5p is associated with cellular viability, migration and apoptosis in renal cell carcinoma
    Peijie Chen, Canbin Lin, Jing Quan, Yulin Lai, Tao He, Liang Zhou, Xiang Pan, Xueling Wu, Yong Wang, Liangchao Ni, Shangqi Yang, Tao Wang, Yongqing Lai
    Molecular Medicine Reports.2017; 16(4): 5023.     CrossRef
  • miR‑660‑5p is associated with cell migration, invasion, proliferation and apoptosis in renal cell carcinoma
    Tao He, Peijie Chen, Lu Jin, Jia Hu, Yifan Li, Liang Zhou, Shangqi Yang, Xiangming Mao, Yaoting Gui, Yun Chen, Yongqing Lai
    Molecular Medicine Reports.2017;[Epub]     CrossRef
  • Synergistic Survival: A New Phenomenon Connected to Adverse Events of First-Line Sunitinib Treatment in Advanced Renal Cell Carcinoma
    Krisztián Nagyiványi, Barna Budai, Krisztina Bíró, Fruzsina Gyergyay, László Noszek, Zsófia Küronya, Hajnalka Németh, Péter Nagy, Lajos Géczi
    Clinical Genitourinary Cancer.2016; 14(4): 314.     CrossRef
  • BSA and ABCB1 polymorphism affect the pharmacokinetics of sunitinib and its active metabolite in Asian mRCC patients receiving an attenuated sunitinib dosing regimen
    Jung-woo Chae, Yi Ling Teo, Han Kiat Ho, Jaeyeon Lee, Hyun-moon Back, Hwi-yeol Yun, Mats O. Karlsson, Kwang-il Kwon, Alexandre Chan
    Cancer Chemotherapy and Pharmacology.2016; 78(3): 623.     CrossRef
  • Key predictive factors for efficacy of axitinib in first-line metastatic renal cell carcinoma: subgroup analysis in Japanese patients from a randomized, double-blind phase II study
    Yoshihiko Tomita, Satoshi Fukasawa, Mototsugu Oya, Hirotsugu Uemura, Nobuo Shinohara, Tomonori Habuchi, Brian I. Rini, Ying Chen, Angel H. Bair, Seiichiro Ozono, Seiji Naito, Hideyuki Akaza
    Japanese Journal of Clinical Oncology.2016; 46(11): 1031.     CrossRef
  • Association Study of a Functional Variant on ABCG2 Gene with Sunitinib-Induced Severe Adverse Drug Reaction
    Siew-Kee Low, Koya Fukunaga, Atsushi Takahashi, Koichi Matsuda, Fumiya Hongo, Hiroyuki Nakanishi, Hiroshi Kitamura, Takamitsu Inoue, Yoichiro Kato, Yoshihiko Tomita, Satoshi Fukasawa, Tomoaki Tanaka, Kazuo Nishimura, Hirotsugu Uemura, Isao Hara, Masato Fu
    PLOS ONE.2016; 11(2): e0148177.     CrossRef
  • Sunitinib therapy for metastatic renal cell carcinoma: A urologist’s perspective
    M Darrad, R Wilson
    Journal of Clinical Urology.2016; 9(1): 32.     CrossRef
  • Sunitinib treatment in patients with advanced renal cell cancer: the Brazilian National Cancer Institute (INCA) experience
    Rafael Corrêa Coelho, Tomás Reinert, Franz Campos, Fábio Affonso Peixoto, Carlos Augusto de Andrade, Thalita Castro, Daniel Herchenhorn
    International braz j urol.2016; 42(4): 694.     CrossRef
  • Tumor suppressor miR-149-5p is associated with cellular migration, proliferation and apoptosis in renal cell carcinoma
    LU JIN, YIFAN LI, JIAJU LIU, SHANGQI YANG, YAOTING GUI, XIANGMING MAO, GUOHUI NIE, YONGQING LAI
    Molecular Medicine Reports.2016; 13(6): 5386.     CrossRef
  • Efficacy and Safety of an Attenuated-Dose Sunitinib Regimen in Metastatic Renal Cell Carcinoma: Results From a Prospective Registry in Singapore
    Hui Shan Tan, Huihua Li, Yu Wen Hong, Chee-Keong Toh, Alvin Wong, Gilberto Lopes, Miah Hiang Tay, Alexandre Chan, Xin Yao, Tiffany Tang, Quan Sing Ng, Ravindran Kanesvaran, Noan Minh Chau, Min-Han Tan
    Clinical Genitourinary Cancer.2015; 13(4): e285.     CrossRef
  • Central and Eastern European Experience with Sunitinib in Metastatic Renal Cell Carcinoma: A Sub-analysis of the Global Expanded-Access Trial
    Eduard Vrdoljak, Lajos Géczi, Jozef Mardiak, Tudor-Eliade Ciuleanu, Sophie Leyman, Ke Zhang, Peter Sajben, Laszlo Torday
    Pathology & Oncology Research.2015; 21(3): 775.     CrossRef
  • Axitinib plasma pharmacokinetics and ethnic differences
    Ying Chen, Akiyuki Suzuki, Michael A. Tortorici, May Garrett, Robert R. LaBadie, Yoshiko Umeyama, Yazdi K. Pithavala
    Investigational New Drugs.2015; 33(2): 521.     CrossRef
  • Management of dose variability and side effects for individualized cancer pharmacotherapy with tyrosine kinase inhibitors
    Tomohiro Terada, Satoshi Noda, Ken-ichi Inui
    Pharmacology & Therapeutics.2015; 152: 125.     CrossRef
  • RandomizEd phase II trial of Sunitinib four weeks on and two weeks off versus Two weeks on and One week off in metastatic clear-cell type REnal cell carcinoma: RESTORE trial
    J.L. Lee, M.K. Kim, I. Park, J.-H. Ahn, D.H. Lee, H.M. Ryoo, C. Song, B. Hong, J.H. Hong, H. Ahn
    Annals of Oncology.2015; 26(11): 2300.     CrossRef
  • Subgroup analysis of Japanese patients in a phase 3 study of lenvatinib in radioiodine‐refractory differentiated thyroid cancer
    Naomi Kiyota, Martin Schlumberger, Kei Muro, Yuichi Ando, Shunji Takahashi, Yasukazu Kawai, Lori Wirth, Bruce Robinson, Steven Sherman, Takuya Suzuki, Katsuki Fujino, Anubha Gupta, Seiichi Hayato, Makoto Tahara
    Cancer Science.2015; 106(12): 1714.     CrossRef
  • Efficacy and Toxicity of Sunitinib in Metastatic Renal Cell Carcinoma Patients in Egypt
    Wael Abdelgawad Edesa, Raafat Ragaey Abdelmalek
    Asian Pacific Journal of Cancer Prevention.2015; 16(5): 1971.     CrossRef
  • Induction of apoptotic cell death by betulin in multidrug-resistant human renal carcinoma cells
    NAM-HUI YIM, YOUNG PIL JUNG, AEYUNG KIM, TAESOO KIM, JIN YEUL MA
    Oncology Reports.2015; 34(2): 1058.     CrossRef
  • Anti-VEGF therapy in mRCC: differences between Asian and non-Asian patients
    Y Wang, T K Choueiri, J-L Lee, M-H Tan, S Y Rha, S A North, C K Kollmannsberger, D F McDermott, D Y C Heng
    British Journal of Cancer.2014; 110(6): 1433.     CrossRef
  • The Effect of ABCG2 Genotype on the Population Pharmacokinetics of Sunitinib in Patients With Renal Cell Carcinoma
    Tomoyuki Mizuno, Masahide Fukudo, Tsuyoshi Fukuda, Tomohiro Terada, Min Dong, Tomomi Kamba, Toshinari Yamasaki, Osamu Ogawa, Toshiya Katsura, Ken-ichi Inui, Alexander A. Vinks, Kazuo Matsubara
    Therapeutic Drug Monitoring.2014; 36(3): 310.     CrossRef
  • Intolerance to Sunitinib Treatment in Hemodialysis Patients With Metastatic Renal Cell Carcinoma
    Ibrahim Yildiz, Fatma Sen, Leyla Kilic, Rumeysa Ciftci, Mert Basaran
    Korean Journal of Urology.2014; 55(1): 74.     CrossRef
  • Impact of adverse events, treatment modifications, and dose intensity on survival among patients with advanced renal cell carcinoma treated with first‐line sunitinib: a medical chart review across ten centers in five European countries
    Camillo Porta, Antonin Levy, Robert Hawkins, Daniel Castellano, Joaquim Bellmunt, Paul Nathan, Ray McDermott, John Wagstaff, Paul Donnellan, John McCaffrey, Francis Vekeman, Maureen P. Neary, Jose Diaz, Faisal Mehmud, Mei Sheng Duh
    Cancer Medicine.2014; 3(6): 1517.     CrossRef
  • Angiogenesis inhibitor therapies for advanced renal cell carcinoma: Toxicity and treatment patterns in clinical practice from a global medical chart review
    WILLIAM K. OH, DAVID McDERMOTT, CAMILLO PORTA, ANTONIN LEVY, REZA ELAIDI, FLORIAN SCOTTE, ROBERT HAWKINS, DANIEL CASTELLANO, JOAQUIM BELLMUNT, SUN YOUNG RHA, JONG-MU SUN, PAUL NATHAN, BRUCE A. FEINBERG, JEFFREY SCOTT, RAY McDERMOTT, JIN-HEE AHN, JOHN WAGS
    International Journal of Oncology.2014; 44(1): 5.     CrossRef
  • Prognostic factors for survival in 1059 patients treated with sunitinib for metastatic renal cell carcinoma
    R J Motzer, B Escudier, R Bukowski, B I Rini, T E Hutson, C H Barrios, X Lin, K Fly, E Matczak, M E Gore
    British Journal of Cancer.2013; 108(12): 2470.     CrossRef
  • Therapy management with sunitinib in patients with metastatic renal cell carcinoma: Key concepts and the impact of clinical biomarkers
    Daniel Castellano, Alain Ravaud, Manuela Schmidinger, Guillermo De Velasco, Federico Vazquez
    Cancer Treatment Reviews.2013; 39(3): 230.     CrossRef
  • Pharmacogenetic determinants associated with sunitinib-induced toxicity and ethnic difference in Korean metastatic renal cell carcinoma patients
    Hye Ryun Kim, Hyung Soon Park, Woo Sun Kwon, Ji Hyun Lee, Yusuke Tanigawara, Sun Min Lim, Hyo Song Kim, Sang Jun Shin, Jung Bae Ahn, Sun Young Rha
    Cancer Chemotherapy and Pharmacology.2013; 72(4): 825.     CrossRef
  • Validation of the MSKCC and Heng Risk Criteria Models for Predicting Survival in Patients with Metastatic Renal Cell Carcinoma Treated with Sunitinib
    Whi-An Kwon, In-Chang Cho, Ami Yu, Byung-Ho Nam, Jae Young Joung, Ho Kyung Seo, Kang Hyun Lee, Jinsoo Chung
    Annals of Surgical Oncology.2013; 20(13): 4397.     CrossRef
  • Efficacy and safety of everolimus in Korean patients with metastatic renal cell carcinoma
    Ki Hyang Kim, Sang Hyun Yoon, Hae-Jung Lee, Hyo Song Kim, Sang Joon Shin, Joong Bae Ahn, Sun Young Rha
    Cancer Chemotherapy and Pharmacology.2013; 72(4): 853.     CrossRef
  • Safety and treatment patterns of angiogenesis inhibitors in patients with advanced renal cell carcinoma in Spain
    Daniel Castellano, Mei Sheng Duh, Caroline Korves, Ellison Dial Suthoff, Maureen Neary, Luis Javier Hernández Pastor, Joaquim Bellmunt
    Expert Opinion on Drug Safety.2013; 12(4): 455.     CrossRef
  • Phase I study of the safety, pharmacokinetics and antitumor activity of famitinib
    Aiping Zhou, Wen Zhang, Chunxiao Chang, Xiaoyan Chen, Dafang Zhong, Qiong Qin, Donghua Lou, Haoyuan Jiang, Jinwan Wang
    Cancer Chemotherapy and Pharmacology.2013; 72(5): 1043.     CrossRef
  • Analysis of the Reasons for Dose Reduction or Discontinuation of Sunitinib in Korean Patients
    노지혜, 이주연, 이혜숙, 김향숙, 강래영, 이용화
    Journal of Korean Society of Health-System Pharmacists.2013; 30(3): 210.     CrossRef
  • Effect of P-Glycoprotein and Breast Cancer Resistance Protein Inhibition on the Pharmacokinetics of Sunitinib in Rats
    Sachiko Kunimatsu, Tomoyuki Mizuno, Masahide Fukudo, Toshiya Katsura
    Drug Metabolism and Disposition.2013; 41(8): 1592.     CrossRef
  • Famitinib in metastatic renal cell carcinoma: a single center study
    Wen ZHANG, Ai-Ping ZHOU, Qiong QIN, Chun-Xiao CHANG, Hao-Yuan JIANG, Jian-Hui MA, Jin-Wan WANG
    Chinese Medical Journal.2013; 126(22): 4277.     CrossRef
  • Management of sunitinib adverse events in renal cell carcinoma patients: The Asian experience
    Aiping ZHOU
    Asia-Pacific Journal of Clinical Oncology.2012; 8(2): 132.     CrossRef
  • Sunitinib as a second-line therapy for advanced GISTs after failure of imatinib: relationship between efficacy and tumor genotype in Korean patients
    Dok Hyun Yoon, Min-Hee Ryu, Baek-Yeol Ryoo, Moyeol Beck, Dae Ro Choi, Yoojin Cho, Jae-Lyun Lee, Heung-Moon Chang, Tae Won Kim, Yoon-Koo Kang
    Investigational New Drugs.2012; 30(2): 819.     CrossRef
  • Current Status of Targeted Therapy for Advanced Renal Cell Carcinoma
    In-Chang Cho, Jinsoo Chung
    Korean Journal of Urology.2012; 53(4): 217.     CrossRef
  • Impact of Genetic Variation in Breast Cancer Resistance Protein (BCRP/ABCG2) on Sunitinib Pharmacokinetics
    Tomoyuki Mizuno, Masahide Fukudo, Tomohiro Terada, Tomomi Kamba, Eijiro Nakamura, Osamu Ogawa, Ken-ichi Inui, Toshiya Katsura
    Drug Metabolism and Pharmacokinetics.2012; 27(6): 631.     CrossRef
  • Response Rates and Adverse Effects of Continuous Once-daily Sunitinib in Patients with Advanced Renal Cell Carcinoma: A Single-center Study in Turkey
    I. Yildiz, F. Sen, M. Basaran, M. Ekenel, F. Agaoglu, E. Darendeliler, H. M. Tunc, F. Ozcan, S. Bavbek
    Japanese Journal of Clinical Oncology.2011; 41(12): 1380.     CrossRef
  • Key predictive factors of axitinib (AG-013736)-induced proteinuria and efficacy: A phase II study in Japanese patients with cytokine-refractory metastatic renal cell Carcinoma
    Yoshihiko Tomita, Hirotsugu Uemura, Hiroyuki Fujimoto, Hiro-omi Kanayama, Nobuo Shinohara, Hayakazu Nakazawa, Keiji Imai, Yoshiko Umeyama, Seiichiro Ozono, Seiji Naito, Hideyuki Akaza
    European Journal of Cancer.2011; 47(17): 2592.     CrossRef
  • Efficacy and Safety of Sunitinib on Metastatic Renal Cell Carcinoma: A Single-Institution Experience
    Eugene Hwang, Hyo Jin Lee, Chong Koo Sul, Jae Sung Lim
    Korean Journal of Urology.2010; 51(7): 450.     CrossRef
  • Kidney Cancer Working Group Report
    S. Naito, Y. Tomita, S. Y. Rha, H. Uemura, M. Oya, H. Z. Song, L. H. Zhong, M. I. B. A. Wahid
    Japanese Journal of Clinical Oncology.2010; 40(Supplement): i51.     CrossRef
  • Hematologic Toxicity in Patients Treated with Sunitinib for Advanced Thyroid Cancer
    Anastasios Gkountouvas, Ifigeneia Kostoglou-Athanassiou, Eirini Veniou, Panagiotis Repousis, Nikolaos Ziras, Philippos Kaldrimidis
    Thyroid.2010; 20(6): 597.     CrossRef
  • The Efficacy and Safety of Sunitinib in Korean Patients with Advanced Renal Cell Carcinoma: High Incidence of Toxicity Leads to Frequent Dose Reduction
    C. Yoo, J. E. Kim, J.-L. Lee, J.-H. Ahn, D. H. Lee, J.-S. Lee, S. Na, C.-S. Kim, J. H. Hong, B. Hong, C. Song, H. Ahn
    Japanese Journal of Clinical Oncology.2010; 40(10): 980.     CrossRef
  • Novel Sunitinib Strategy in Metastatic Renal Cell Carcinoma on Hemodialysis: Intermittent Dose of Sunitinib after Hemodialysis
    Sang Hyun Yoon, Ki Hyang Kim, Junjeong Choi, Gun Min Kim, Joo Hoon Kim, Hyo Song Kim, Young Nyun Park, Sun Young Rha
    Cancer Research and Treatment.2010; 42(3): 180.     CrossRef
  • Comparative Analysis between Immunochemotherapy and Target Therapy for Metastatic Renal Cell Carcinoma: Overview of Treatment-Related Adverse Events and the Dropout Rate in Korea
    Jee Han Lee, Sung-Goo Chang, Seung Hyun Jeon, Gyeong Eun Min, Koo Han Yoo
    Korean Journal of Urology.2010; 51(6): 379.     CrossRef
  • 12,625 View
  • 74 Download
  • 57 Crossref
Close layer

Cancer Res Treat : Cancer Research and Treatment
Close layer
TOP