Skip Navigation
Skip to contents

Cancer Res Treat : Cancer Research and Treatment

OPEN ACCESS

Search

Page Path
HOME > Search
18 "Phase I"
Filter
Filter
Article category
Keywords
Publication year
Authors
Funded articles
Original Articles
Breast cancer
A Phase II Trial of S-1 and Oxaliplatin in Patients with Metastatic Breast Cancer Previously Treated with Anthracycline and Taxane (KCSG-BR07-03)
Dae-Won Lee, Bhumsuk Keam, Keun Seok Lee, Jin-Hee Ahn, Joohyuk Sohn, Jin Seok Ahn, Moon Hee Lee, Jee Hyun Kim, Kyung Eun Lee, Hyo Jung Kim, Si-Young Kim, Yeon Hee Park, Chan-Young Ock, Kyung-Hun Lee, Sae-Won Han, Sung-Bae Kim, Young Hyuck Im, Hyun Cheol Chung, Do-Youn Oh, Seock-Ah Im
Cancer Res Treat. 2023;55(2):523-530.   Published online November 8, 2022
DOI: https://doi.org/10.4143/crt.2022.1360
AbstractAbstract PDFPubReaderePub
Purpose
This single-arm phase II trial investigate the efficacy and safety of S-1 plus oxaliplatin (SOX) in patients with metastatic breast cancer.
Materials and Methods
Patients with metastatic breast cancer previously treated with anthracyclines and taxanes were enrolled. Patients received S-1 (40-60 mg depending on patient’s body surface area, twice a day, day 1-14) and oxaliplatin (130 mg/m2, day 1) in 3 weeks cycle until disease progression or unacceptable toxicity. The primary endpoint was objective response rate (ORR) according to Response Evaluation Criteria in Solid Tumor 1.1. Secondary endpoints included time-to-progression (TTP), duration-of-response (DoR), overall survival (OS), and adverse events.
Results
A total of 87 patients were enrolled from 11 institutions in Korea. Hormone receptor was positive in 54 (62.1%) patients and six (6.9%) had human epidermal growth factor receptor 2–positive disease. Forty-eight patients (85.1%) had visceral metastasis and 74 (55.2%) had more than three sites of metastases. The ORR of SOX regimen was 38.5% (95% confidence interval [CI], 26.9 to 50.0) with a median TTP of 6.0 months (95% CI, 5.1 to 6.9). Median DoR and OS were 10.3 months (95% CI, 5.5 to 15.1) and 19.4 (95% CI, not estimated) months, respectively. Grade 3 or 4 neutropenia was reported in 28 patients (32.1%) and thrombocytopenia was observed in 23 patients (26.6%).
Conclusion
This phase II study showed that SOX regimen is a reasonable option in metastatic breast cancer previously treated with anthracyclines and taxanes.

Citations

Citations to this article as recorded by  
  • Unraveling the immune landscape and therapeutic biomarker PMEPA1 for oxaliplatin resistance in colorectal cancer: A comprehensive approach
    Zhengguang Zhang, Tianming Lu, Zhe Zhang, Zixian Liu, Ruoning Qian, Ruogu Qi, Fuqiong Zhou, Min Li
    Biochemical Pharmacology.2024; 222: 116117.     CrossRef
  • Efficacy and safety of utidelone plus capecitabine in advanced first-line therapy for metastatic breast cancer: A multicenter real-world study
    Pingping Bi, Xi Wang, Rui Liu, Xiuqin Li, Shanrong Wei, Jiawen Zhao, Xin Tan, Fan Zhang, Qing Mao, Ying Zhang, Baoyan Tang, Xueqiong Xun, Rong Guo, Kai Zheng, Shaoqiang Zhou, Shicong Tang
    Surgery Open Science.2023; 16: 171.     CrossRef
  • 4,563 View
  • 165 Download
  • 2 Web of Science
  • 2 Crossref
Close layer
Sarcoma
Whole-Genome and Transcriptome Sequencing Identified NOTCH2 and HES1 as Potential Markers of Response to Imatinib in Desmoid Tumor (Aggressive Fibromatosis): A Phase II Trial Study
Joonha Kwon, Jun Hyeong Lee, Young Han Lee, Jeeyun Lee, Jin-Hee Ahn, Se Hyun Kim, Seung Hyun Kim, Tae Il Kim, Kum-Hee Yun, Young Suk Park, Jeong Eun Kim, Kyu Sang Lee, Jung Kyoon Choi, Hyo Song Kim
Cancer Res Treat. 2022;54(4):1240-1255.   Published online January 17, 2022
DOI: https://doi.org/10.4143/crt.2021.1194
AbstractAbstract PDFSupplementary MaterialPubReaderePub
Purpose
Desmoid tumor, also known as aggressive fibromatosis, is well-characterized by abnormal Wnt/β-catenin signaling. Various therapeutic options, including imatinib, are available to treat desmoid tumor. However, the molecular mechanism of why imatinib works remains unclear. Here, we describe potential roles of NOTCH2 and HES1 in clinical response to imatinib at genome and transcriptome levels.
Materials and Methods
We identified somatic mutations in coding and noncoding regions via whole-genome sequencing. To validate the genetic interaction with expression level in desmoid-tumor condition, we utilized large-scale whole-genome sequencing and transcriptome datasets from the Pan-Cancer Analysis of Whole Genomes project. RNA-sequencing was performed using prospective and retrospective cohort samples to evaluate the expressional relevance with clinical response.
Results
Among 20 patients, four (20%) had a partial response and 14 (66.7%) had stable disease, 11 of which continued for ≥ 1 year. With gene-wise functional analyses, we detected a significant correlation between recurrent NOTCH2 noncoding mutations and clinical response to imatinib. Based on Pan-Cancer Analysis of Whole Genomes data analyses, NOTCH2 mutations affect expression levels particularly in the presence of CTNNB1 missense mutations. By analyzing RNA-sequencing with additional desmoid tumor samples, we found that NOTCH2 expression was significantly correlated with HES1 expression. Interestingly, NOTCH2 had no statistical power to discriminate between responders and non-responders. Instead, HES1 was differentially expressed with statistical significance between responders and non-responders.
Conclusion
Imatinib was effective and well tolerated for advanced desmoid tumor treatment. Our results show that HES1, regulated by NOTCH2, as an indicator of sensitivity to imatinib, and an important therapeutic consideration for desmoid tumor.

Citations

Citations to this article as recorded by  
  • The Notch signaling pathway in desmoid tumor: Recent advances and the therapeutic prospects
    Chuanxi Zheng, Jianghong Huang, Gang Xu, Wei Li, Xin Weng, Shiquan Zhang
    Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease.2024; 1870(1): 166907.     CrossRef
  • Long‐term result of 125I seed brachytherapy for pediatric desmoid tumor in the head and neck
    Yi‐Wei Zhong, Xiao‐Ming Lyu, Yan Shi, Chuan‐Bin Guo, Jian‐Guo Zhang, Lei Zheng
    Pediatric Blood & Cancer.2023;[Epub]     CrossRef
  • Update on Familial Adenomatous Polyposis-Associated Desmoid Tumors
    Wanjun Yang, Pei-Rong Ding
    Clinics in Colon and Rectal Surgery.2023; 36(06): 400.     CrossRef
  • Multimodality Imaging Assessment of Desmoid Tumors: The Great Mime in the Era of Multidisciplinary Teams
    Igino Simonetti, Federico Bruno, Roberta Fusco, Carmen Cutolo, Sergio Venanzio Setola, Renato Patrone, Carlo Masciocchi, Pierpaolo Palumbo, Francesco Arrigoni, Carmine Picone, Andrea Belli, Roberta Grassi, Francesca Grassi, Antonio Barile, Francesco Izzo,
    Journal of Personalized Medicine.2022; 12(7): 1153.     CrossRef
  • 6,670 View
  • 270 Download
  • 4 Web of Science
  • 4 Crossref
Close layer
Breast Cancer
Talazoparib Versus Chemotherapy in Patients with HER2-negative Advanced Breast Cancer and a Germline BRCA1/2 Mutation Enrolled in Asian Countries: Exploratory Subgroup Analysis of the Phase III EMBRACA Trial
Kyung-Hun Lee, Joohyuk Sohn, Annabel Goodwin, Tiziana Usari, Silvana Lanzalone, Seock-Ah Im, Sung-Bae Kim
Cancer Res Treat. 2021;53(4):1084-1095.   Published online March 24, 2021
DOI: https://doi.org/10.4143/crt.2020.1381
AbstractAbstract PDFSupplementary MaterialPubReaderePub
Purpose
We evaluated study outcomes in patients enrolled in Asian regions in the phase III EMBRACA trial of talazoparib vs. chemotherapy.
Materials and Methods
Patients with human epidermal growth factor receptor 2–negative germline BRCA1/2-mutated advanced breast cancer who received prior chemotherapy were randomized 2:1 to talazoparib 1 mg/day or chemotherapy (physician’s choice). Primary endpoint was progression-free survival (PFS) per independent central review in the intent-to-treat (ITT) population. This post-hoc analysis evaluated efficacy/safety endpoints in the ITT population of patients enrolled in Asian regions.
Results
Thirty-three patients were enrolled at Asian sites (talazoparib, n=23; chemotherapy, n=10). Baseline characteristics were generally comparable with the overall EMBRACA population. In Asian patients, median PFS was 9.0 months (95% confidence interval [CI] 3.0, 15.2) for talazoparib and 7.1 months (95% CI, 1.2, not reached) for chemotherapy (hazard ratio [HR] 0.74 [95% CI, 0.22, 2.44]). Objective response rate was numerically higher for talazoparib vs. chemotherapy (62.5% [95% CI, 35.4, 84.8] vs. 25.0% [95% CI, 3.2, 65.1]). Median overall survival was 20.7 months (95% CI, 9.4, 40.1) versus 21.2 months (95% CI, 2.7, 35.0) months (HR, 1.41 [95% CI, 0.49, 4.05]). In Asian patients, fewer grade 3/4 adverse events (AEs), serious AEs (SAEs), grade 3/4 SAEs, and AEs resulting in dose reduction/discontinuation occurred with talazoparib than chemotherapy; for talazoparib, the frequency of these events was lower in Asian patients versus overall EMBRACA population.
Conclusion
In this subgroup analysis, talazoparib numerically improved efficacy versus chemotherapy and was generally well tolerated in Asian patients, with fewer grade 3/4 TEAEs, SAEs, and TEAEs leading to dose modification vs. the overall EMBRACA population.

Citations

Citations to this article as recorded by  
  • HR-positive/HER2-negative breast cancer arising in patients with or without BRCA2 mutation: different biological phenotype and similar prognosis
    Pu-Chun Li, Yi-Fan Zhu, Jia-Ni Pan, Qiao-Yan Zhu, Yu-Yang Liao, Xiao-Wen Ding, Lin-Feng Zheng, Wen-Ming Cao
    Therapeutic Advances in Medical Oncology.2024;[Epub]     CrossRef
  • Detection and analysis of the safety profile of talazoparib based on FAERS database
    Mufei Tang, Peiyan Liu, Linzhe Du, Yuanyuan Li, Jinjin Chen, Yang Li
    Expert Opinion on Drug Safety.2024; : 1.     CrossRef
  • Facing inevitable PARPis resistance: Mechanisms and therapeutic strategies for breast cancer treatment
    Haixia Liu, Xiaohui Chen, Yimin Jia, Hengyi Chen, Xiaohui Wang, Guoxiang Liu, Yang Luo
    Interdisciplinary Medicine.2023;[Epub]     CrossRef
  • Sustained delivery of PARP inhibitor Talazoparib for the treatment of BRCA-deficient ovarian cancer
    Shicheng Yang, Allen Green, Needa Brown, Alexis Robinson, Merline Senat, Bryanna Testino, Daniela M. Dinulescu, Srinivas Sridhar
    Frontiers in Oncology.2023;[Epub]     CrossRef
  • Pan-Asian adapted ESMO Clinical Practice Guidelines for the diagnosis, staging and treatment of patients with metastatic breast cancer
    S.-A. Im, A. Gennari, Y.H. Park, J.H. Kim, Z.-F. Jiang, S. Gupta, T.H. Fadjari, K. Tamura, M.Y. Mastura, M.L.T. Abesamis-Tiambeng, E.H. Lim, C.-H. Lin, A. Sookprasert, N. Parinyanitikul, L.-M. Tseng, S.-C. Lee, P. Caguioa, M. Singh, Y. Naito, R.A. Hukom,
    ESMO Open.2023; 8(3): 101541.     CrossRef
  • Molecular Biology Mechanisms and Emerging Therapeutics of Triple-Negative Breast Cancer
    Zhiying Zhang, Rui Zhang, Donghai Li
    Biologics: Targets and Therapy.2023; Volume 17: 113.     CrossRef
  • Development of the PARP inhibitor talazoparib for the treatment of advanced BRCA1 and BRCA2 mutated breast cancer
    Evthokia A. Hobbs, Jennifer K. Litton, Timothy A. Yap
    Expert Opinion on Pharmacotherapy.2021; : 1.     CrossRef
  • 8,129 View
  • 265 Download
  • 6 Web of Science
  • 7 Crossref
Close layer
Gastrointestinal cancer
Targeting Hypoxia Using Evofosfamide and Companion Hypoxia Imaging of FMISO-PET in Advanced Biliary Tract Cancer
Jeesun Yoon, Seo Young Kang, Kyung-Hun Lee, Gi Jeong Cheon, Do-Youn Oh
Cancer Res Treat. 2021;53(2):471-479.   Published online October 22, 2020
DOI: https://doi.org/10.4143/crt.2020.577
AbstractAbstract PDFSupplementary MaterialPubReaderePub
Purpose
Hypoxia is widely known as one of the mechanisms of chemoresistance and as an environmental condition which triggers invasion and metastasis of cancer. Evofosfamide is a hypoxia-activated prodrug of the cytotoxin bromo-isophosphoramide mustard conjugated with 2-nitroimidazole. Biliary tract cancer (BTC) is known to contain large hypoxic area. This study evaluated the efficacy and safety of evofosfamide as a second-line treatment of advanced BTC.
Materials and Methods
Patients received evofosfamide at a dose of 340 mg/m2 on days 1, 8, and 15 of every 28-day cycle. Primary end-point was progression-free survival (PFS) rate at 4-months (4m-PFSR). Secondary end-points included overall survival (OS), PFS, disease control rate (DCR), metabolic response by 18F-fluorodeoxyglucose positron emission tomography (PET), hypoxic parameters evaluated by 18F-fluoromisonidazole (FMISO) PET and toxicity.
Results
Twenty patients were treated with evofosfamide, with 16 response-evaluable patients. There was no objective response; stable disease was observed in nine patients, with a DCR of 56.25%. 4m-PFSR was 40.6%. Median PFS was 3.60 months (95% confidence interval [CI], 1.68 to 5.52). Median OS was 6.37 months (95% CI, 3.94 to 8.79). Reduction of tumor metabolic activity was observed in eight of 15 patients (53.3%). High baseline hypoxic parameters were associated with poor PFS. Change of hypoxic parameters between pretreatment and post-treatment reflected hypoxic-activated drug response. There was no treatment-related death.
Conclusion
Evofosfamide as second-line treatment of advanced BTC showed acceptable safety and comparable efficacy to other agents. Changes in volumetric parameters measured with FMISO PET, showing the degree of tumor hypoxia, reflected the response to evofosfamide based on the mode of action.

Citations

Citations to this article as recorded by  
  • Functional Imaging of Hypoxia: PET and MRI
    Ryan C. Perez, DaeHee Kim, Aaron W. P. Maxwell, Juan C. Camacho
    Cancers.2023; 15(13): 3336.     CrossRef
  • Phase Ib dose-escalation study of the hypoxia-modifier Myo-inositol trispyrophosphate in patients with hepatopancreatobiliary tumors
    Marcel A. Schneider, Michael Linecker, Ralph Fritsch, Urs J. Muehlematter, Daniel Stocker, Bernhard Pestalozzi, Panagiotis Samaras, Alexander Jetter, Philipp Kron, Henrik Petrowsky, Claude Nicolau, Jean-Marie Lehn, Bostjan Humar, Rolf Graf, Pierre-Alain C
    Nature Communications.2021;[Epub]     CrossRef
  • 5,663 View
  • 180 Download
  • 2 Web of Science
  • 2 Crossref
Close layer
Osimertinib in Patients with T790M-Positive Advanced Non-small Cell Lung Cancer: Korean Subgroup Analysis from Phase II Studies
Myung-Ju Ahn, Ji-Youn Han, Dong-Wan Kim, Byoung Chul Cho, Jin-Hyoung Kang, Sang-We Kim, James Chih-Hsin Yang, Tetsuya Mitsudomi, Jong Seok Lee
Cancer Res Treat. 2020;52(1):284-291.   Published online July 23, 2019
DOI: https://doi.org/10.4143/crt.2019.200
AbstractAbstract PDFPubReaderePub
Purpose
Osimertinib is a third-generation, irreversible, oral epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) that potently and selectively inhibits both EGFR sensitizing mutation and EGFR T790M and has demonstrated efficacy in non-small cell lung cancer (NSCLC) central nervous system (CNS) metastases. We present results of a subgroup analysis of Korean patients from the pooled data of two global phase II trials: AURA extension (NCT01802632) and AURA2 (NCT02094261).
Materials and Methods
Enrolled patients had EGFR T790M-positive NSCLC and disease progression during or after EGFR-TKI therapy. Patients received osimertinib 80 mg orally once daily until disease progression. The primary endpoint was objective response rate (ORR).
Results
In total, 66 Korean patients received osimertinib treatment with a median treatment duration of 19 months. In the evaluable-for-response population (n=62), ORR was 74% (95% confidence interval [CI], 61.5 to 84.5) and median duration of response was 9.8 months (95% CI, 7.1 to 16.8). In the full analysis set (n=66), median progression-free survival was 10.9 months (95% CI, 8.3 to 15.0; data cutoff November 1, 2016), and median overall survival was 29.2 months (95% CI, 24.8 to 35.7; data cutoff May 1, 2018). Eight patients with CNS metastases were evaluable for response, none of whom showed CNS progression. The most common adverse events were rash (53%), cough (33%), paronychia, diarrhea, and decreased appetite (each 32%).
Conclusion
Efficacy and safety profiles of osimertinib in this subgroup are consistent with the global phase II pooled population, which supports osimertinib as a recommended treatment for Korean patients with T790M positive NSCLC.

Citations

Citations to this article as recorded by  
  • Pan-Asian adapted ESMO Clinical Practice Guidelines for the diagnosis, treatment and follow-up of patients with oncogene-addicted metastatic non-small-cell lung cancer
    S.-H. Lee, J. Menis, T.M. Kim, H.R. Kim, C. Zhou, S.A. Kurniawati, K. Prabhash, H. Hayashi, D.D.-W. Lee, M.S. Imasa, Y.L. Teh, J.C.-H. Yang, T. Reungwetwattana, V. Sriuranpong, C.-E. Wu, Y. Ang, M. Sabando, M. Thiagarajan, H. Mizugaki, V. Noronha, M. Yuli
    ESMO Open.2024; 9(12): 103996.     CrossRef
  • The role of brain radiotherapy for EGFR- and ALK-positive non-small-cell lung cancer with brain metastases: a review
    Valerio Nardone, Caterina Romeo, Emma D’Ippolito, Pierpaolo Pastina, Maria D’Apolito, Luigi Pirtoli, Michele Caraglia, Luciano Mutti, Giovanna Bianco, Antonella Consuelo Falzea, Rocco Giannicola, Antonio Giordano, Pierosandro Tagliaferri, Claudia Vincigue
    La radiologia medica.2023; 128(3): 316.     CrossRef
  • The Relationship Between Short-Term Surrogate Endpoint Indicators and mPFS and mOS in Clinical Trials of Malignant Tumors: A Case Study of Approved Molecular Targeted Drugs for Non-Small-Cell Lung Cancer in China
    Mingjun Rui, Zijing Wang, Zhengyang Fei, Yao Wu, Yingcheng Wang, Lei Sun, Ye Shang, Hongchao Li
    Frontiers in Pharmacology.2022;[Epub]     CrossRef
  • Efficacy and Safety of SH-1028 in Patients With EGFR T790M-Positive NSCLC: A Multicenter, Single-Arm, Open-Label, Phase 2 Trial
    Anwen Xiong, Shengxiang Ren, Huaimin Liu, Liyun Miao, Lei Wang, Jianhua Chen, Wei Li, Runpu Li, Xiang Wang, Zhiwei Lu, Donglin Wang, Xiaohong Wu, Zhihua Liu, Ligang Xing, Yimin Mao, Chunling Liu, Aiping Zeng, Hongrui Niu, Yingying Du, Yuping Sun, Yueyin P
    Journal of Thoracic Oncology.2022; 17(10): 1216.     CrossRef
  • Treatment of Brain Metastases of Non-Small Cell Lung Carcinoma
    Agnieszka Rybarczyk-Kasiuchnicz, Rodryg Ramlau, Katarzyna Stencel
    International Journal of Molecular Sciences.2021; 22(2): 593.     CrossRef
  • 9,244 View
  • 297 Download
  • 5 Web of Science
  • 5 Crossref
Close layer
Pazopanib for the Treatment of Non-clear Cell Renal Cell Carcinoma: A Single-Arm, Open-Label, Multicenter, Phase II Study
Ki Sun Jung, Su Jin Lee, Se Hoon Park, Jae-Lyun Lee, Se-Hoon Lee, Jae Yun Lim, Jung Hun Kang, Suee Lee, Sun Young Rha, Kyung Hee Lee, Ho Young Kim, Ho Yeong Lim
Cancer Res Treat. 2018;50(2):488-494.   Published online May 22, 2017
DOI: https://doi.org/10.4143/crt.2016.584
AbstractAbstract PDFSupplementary MaterialPubReaderePub
Purpose
The optimal treatment strategy for patients with metastatic non-clear cell type renal cell carcinoma (nccRCC) remains unclear. Although several inhibitors of vascular endothelial growth factor have recently shown efficacy against nccRCC, the clinical benefit of pazopanib in nccRCC has not been analyzed. We therefore designed a single-arm, open-label, phase II study to determine the efficacy and safety of pazopanib in patients with nccRCC.
Materials and Methods
Patientswith locally advanced or metastatic nccRCC, exceptfor collecting duct or sarcomatoid type, received 800 mg/day of pazopanib daily until progression of disease or intolerable toxicity. One cyclewas defined as 4weeks and tumorresponsewas evaluated every two cycles. The primary objective was overall response rate (ORR).
Results
A total of 29 eligible patients were enrolled at nine centers in Korea from December 2012 and September 2014. The median age of the patients was 58 years (range, 27 to 76 years) and 21 patients (72%) were male. Regarding histology type, 19 patients had papillary, three had chromophobe, two had unclassified and five had unknown non-clear cell type. Of 28 evaluable patients, eight achieved a confirmed partial response with ORR of 28%. The median progression-free survival was 16.5 months (95% confidence interval, 10.9 to 22.1) and median overall survival was not reached. Sixteen patients (55%) experienced treatment-related toxicity of grade 3 or more, but most adverse events were overcome through dose reduction and delay.
Conclusion
In this prospective phase II study, pazopanib demonstrated promising activity and tolerable safety profile in patients with metastatic nccRCC.

Citations

Citations to this article as recorded by  
  • Advances in non‐clear cell renal cell carcinoma management: From heterogeneous biology to treatment options
    Nathaniel R. Wilson, Yusuf Acikgoz, Elshad Hasanov
    International Journal of Cancer.2024; 154(6): 947.     CrossRef
  • Advanced renal cell carcinoma management: the Latin American Cooperative Oncology Group (LACOG) and the Latin American Renal Cancer Group (LARCG) consensus update
    Andrey Soares, Fernando Sabino Marques Monteiro, Karine Martins da Trindade, Adriano Gonçalves e Silva, Ana Paula Garcia Cardoso, André Deeke Sasse, André P. Fay, André Paternò Castello Dias Carneiro, Antonio Machado Alencar Junior, Augusto César de Andra
    Journal of Cancer Research and Clinical Oncology.2024;[Epub]     CrossRef
  • Papillary Renal Cell Carcinoma: Current Evidence and Future Directions
    Albert Jang, Charbel S. Hobeika, Shilpa Gupta
    Kidney Cancer.2024; 8(1): 61.     CrossRef
  • Comparison of the efficacy of sunitinib and pazopanib in patients with advanced non-clear renal cell carcinoma
    Hasan Cagri Yildirim, Ertuğrul Bayram, Elvin Chalabiyev, Nargız Majidova, Tugay Avci, Halil Göksel Güzel, Caner Kapar, Mehmet Uzun, Perihan Perkin, Fahri Akgül, Saadet Sim Yildirim, Seda Sali, Anil Yildiz, Seher Nazli Kazaz, Engin Hendem, Murat Arcagok, G
    Journal of Chemotherapy.2024; : 1.     CrossRef
  • Real World Data of Diagnosis, Survival, and Treatment Outcomes in Patients With Metastatic Non Clear Cell Renal Cell Carcinoma
    Floriane Izarn, Benoît Allignet, Romane Gille, Helen Boyle, Eve-Marie Neidhardt, Sylvie Négrier, Aude Fléchon
    Clinical Genitourinary Cancer.2023; 21(2): e35.     CrossRef
  • SEOM SOGUG clinical guideline for treatment of kidney cancer (2022)
    María José Méndez-Vidal, Martin Lázaro Quintela, Nuria Lainez-Milagro, Begoña Perez-Valderrama, Cristina Suárez Rodriguez, José Ángel Arranz Arija, Ignacio Peláez Fernández, Enrique Gallardo Díaz, Julio Lambea Sorrosal, Aránzazu González-del-Alba
    Clinical and Translational Oncology.2023; 25(9): 2732.     CrossRef
  • Clinical Features and Prognostic Factors of Metastatic Non-Clear Cell Renal Cell Carcinoma: A Multicenter Study from the Turkish Oncology Group Kidney Cancer Consortium
    Cihan Erol, Emre Yekedüz, Deniz Tural, Serdar Karakaya, Nihan Şentürk Öztaş, Gökhan Uçar, Saadettin Kılıçkap, İsmail Ertürk, Özlem Nuray Sever, Çağatay Arslan, Ahmet Küçükarda, Orçun Can, Özlem Balvan, Satı Coşkun Yazgan, Mustafa Özgüroğlu, Berna Öksüzoğl
    Urologia Internationalis.2023; 107(6): 595.     CrossRef
  • The efficacy of molecular targeted therapy and nivolumab therapy for metastatic non‐clear cell renal cell carcinoma: A retrospective analysis using the Michinoku Japan urological cancer study group database
    Tomoyuki Koguchi, Sei Naito, Shingo Hatakeyama, Kazuyuki Numakura, Yumina Muto, Renpei Kato, Takahiro Kojima, Yoshihide Kawasaki, Kento Morozumi, Shuya Kandori, Sadafumi Kawamura, Hiroyuki Nishiyama, Akihiro Ito, Tomonori Habuchi, Wataru Obara, Chikara Oh
    Cancer Medicine.2023; 12(22): 20677.     CrossRef
  • Kidney Cancer, Version 3.2022, NCCN Clinical Practice Guidelines in Oncology
    Robert J. Motzer, Eric Jonasch, Neeraj Agarwal, Ajjai Alva, Michael Baine, Kathryn Beckermann, Maria I. Carlo, Toni K. Choueiri, Brian A. Costello, Ithaar H. Derweesh, Arpita Desai, Yasser Ged, Saby George, John L. Gore, Naomi Haas, Steven L. Hancock, Pay
    Journal of the National Comprehensive Cancer Network.2022; 20(1): 71.     CrossRef
  • Systematic Review of Treatment of Metastatic Non-Clear Cell Renal Cell Carcinoma
    Jason R. Brown, Adam Calaway, Erik Castle, Jorge Garcia, Pedro C. Barata
    Kidney Cancer.2022; 6(1): 53.     CrossRef
  • Non-Clear Cell Renal Cell Carcinoma: Molecular Pathogenesis, Innovative Modeling, and Targeted Therapeutic Approaches
    Niloofar Khoshdel Rad, Maryam Vahidyeganeh, Mahsa Mohammadi, Anastasia Shpichka, Peter Timashev, Nikoo Hossein-Khannazer, Massoud Vosough
    International Journal of Translational Medicine.2022; 2(4): 555.     CrossRef
  • Systemic Therapies for the Management of Non–Clear Cell Renal Cell Carcinoma: What Works, What Doesn’t, and What the Future Holds
    Panagiotis Zoumpourlis, Giannicola Genovese, Nizar M. Tannir, Pavlos Msaouel
    Clinical Genitourinary Cancer.2021; 19(2): 103.     CrossRef
  • Comprehensive review of chromophobe renal cell carcinoma
    Rohan Garje, Dean Elhag, Hesham A Yasin, Luna Acharya, Daniel Vaena, Laila Dahmoush
    Critical Reviews in Oncology/Hematology.2021; 160: 103287.     CrossRef
  • A Systematic Review of Systemic Treatment Options for Advanced Non-Clear Cell Renal Cell Carcinoma
    Chelsea K. Osterman, Tracy L. Rose
    Kidney Cancer.2020; 4(1): 15.     CrossRef
  • Occurrence of abscesses during treatment with pazopanib in metastatic renal cancer: a case report
    Ivana Puliafito, Alessio Russo, Dorotea Sciacca, Caterina Puglisi, Dario Giuffrida
    Journal of Medical Case Reports.2020;[Epub]     CrossRef
  • On the Shoulders of Giants: The Evolution of Renal Cell Carcinoma Treatment—Cytokines, Targeted Therapy, and Immunotherapy
    Janice P. Dutcher, Ronan Flippot, Jaleh Fallah, Bernard Escudier
    American Society of Clinical Oncology Educational Book.2020; (40): 418.     CrossRef
  • Pazopanib as a possible option for the treatment of metastatic non-clear cell renal carcinoma patients: a systematic review
    Melissa Bersanelli, Matteo Brunelli, Letizia Gnetti, Umberto Maestroni, Sebastiano Buti
    Therapeutic Advances in Medical Oncology.2020;[Epub]     CrossRef
  • Management of Metastatic Renal Cell Carcinoma with Variant Histologies
    Ronan Flippot, Vijay Damarla, Bradley A. McGregor
    Urologic Clinics of North America.2020; 47(3): 319.     CrossRef
  • Discovery of Pyrido[3′,2′:5,6]thiopyrano[4,3-d]pyrimidine-Based Antiproliferative Multikinase Inhibitors
    Silvia Salerno, Elisabetta Barresi, Aída Nelly García-Argáez, Sabrina Taliani, Francesca Simorini, Giorgio Amendola, Stefano Tomassi, Sandro Cosconati, Ettore Novellino, Federico Da Settimo, Anna Maria Marini, Lisa Dalla Via
    ACS Medicinal Chemistry Letters.2019; 10(4): 457.     CrossRef
  • Cabozantinib in advanced non-clear-cell renal cell carcinoma: a multicentre, retrospective, cohort study
    Nieves Martínez Chanzá, Wanling Xie, Mehmet Asim Bilen, Hannah Dzimitrowicz, Jarred Burkart, Daniel M Geynisman, Archana Balakrishnan, I Alex Bowman, Rohit Jain, Walter Stadler, Yousef Zakharia, Vivek Narayan, Benoit Beuselinck, Rana R McKay, Abhishek Tri
    The Lancet Oncology.2019; 20(4): 581.     CrossRef
  • Recent advances in the systemic treatment of metastatic non‐clear cell renal cell carcinomas
    Keiichi Ito
    International Journal of Urology.2019; 26(9): 868.     CrossRef
  • Long-term response of metastatic hereditary leiomyomatosis and renal cell carcinoma syndrome associated renal cell carcinoma to bevacizumab plus erlotinib after temsirolimus and axitinib treatment failures
    Inkeun Park, Young Sup Shim, Heounjeong Go, Bum Sik Hong, Jae Lyun Lee
    BMC Urology.2019;[Epub]     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
  • The Role of Pazopanib in Non-Clear Cell Renal Cell Carcinoma: A Systematic Review
    Gregory T. Sneed, Sukdong Lee, Jamie N. Brown, Julia M. Hammond
    Clinical Genitourinary Cancer.2019; 17(6): 419.     CrossRef
  • A Multicenter Phase II Trial of Axitinib in Patients With Recurrent or Metastatic Non–clear-cell Renal Cell Carcinoma Who Had Failed Prior Treatment With Temsirolimus
    Inkeun Park, Se Hoon Lee, Jae Lyun Lee
    Clinical Genitourinary Cancer.2018; 16(5): e997.     CrossRef
  • Clinical Outcomes in Patients With Metastatic Papillary Renal-Cell Carcinoma: A Multi-Institutional Study in Japan
    Keiichi Ito, Shuji Mikami, Katsunori Tatsugami, Naoya Masumori, Nobuo Shinohara, Tsunenori Kondo, Shotaro Nakanishi, Yoji Nagashima, Masatoshi Eto, Tomomi Kamba, Naoto Kuroda, Yoshihiko Tomita, Hideyasu Matsuyama, Tetsuro Onishi, Tomoyasu Tsushima, Hayaka
    Clinical Genitourinary Cancer.2018; 16(6): e1201.     CrossRef
  • 10,565 View
  • 490 Download
  • 35 Web of Science
  • 26 Crossref
Close layer
An Open-Label, Randomized, Parallel, Phase II Trial to Evaluate the Efficacy and Safety of a Cremophor-Free Polymeric Micelle Formulation of Paclitaxel as First-Line Treatment for Ovarian Cancer: A Korean Gynecologic Oncology Group Study (KGOG-3021)
Shin-Wha Lee, Yong-Man Kim, Chi Heum Cho, Young Tae Kim, Seok Mo Kim, Soo Young Hur, Jae-Hoon Kim, Byoung-Gie Kim, Seung-Cheol Kim, Hee-Sug Ryu, Soon Beom Kang
Cancer Res Treat. 2018;50(1):195-203.   Published online March 21, 2017
DOI: https://doi.org/10.4143/crt.2016.376
AbstractAbstract PDFPubReaderePub
Purpose
Genexol-PM is a biodegradable cremophor EL–free polymeric micelle formulation of paclitaxel. Here,we compared efficacy and safety of Genexol-PM plus carboplatin versus Genexol plus carboplatin for ovarian cancer treatment.
Materials and Methods
In this multicenter, randomized, phase II study, patients with International Federation of Gynecology and Obstetrics IC-IV epithelial ovarian cancer were randomly assigned (1:1) to receive Genexol-PM 260 mg/m2 or Genexol 175 mg/m2 with 5 area under the curve carboplatin every 3weeks (6 cycles). The primary endpointwas the carbohydrate antigen 125 and Response Evaluation Criteria In Solid Tumor composite overall response rate (ORR).
Results
Of 131 enrolled patients, 98 were included in intention-to-treat analysis. Mean dosages were 260.00±0.00 mg/m2 Genexol-PM or 174.24±3.81 mg/m2 Genexol. Median followup was 18.0 months (range, 6.1 to 33.8 months). ORR was 88.0% (95% confidence interval [CI], 80.4 to 95.6) with Genexol-PM, and 77.1% (95% CI, 67.1 to 87.1) with Genexol (noninferiority threshold, 16.3%). Median time to progression was 14.8 months (95% CI, 11.3 to 20.2) with Genexol-PM and 15.4 months (95% CI, 13.2 to 29.6) with Genexol (p=0.550). Overall, six patients died. Neutropenia was the most common toxicity (incidences of 86.0% vs. 77.1%, p=0.120). Peripheral neuropathy incidences were 84.0% versus 64.6% (p= 0.148). Peripheral neuropathy of ≥ grade 3 occurred in one patient receiving Genexol. All toxicities were manageable.
Conclusion
Genexol-PM plus carboplatin as first-line treatment in patients with epithelial ovarian cancer demonstrated non-inferior efficacy and well-tolerated toxicities compared with the standard paclitaxel regimen. Further studies are warranted to optimize the dose and schedule, and to investigate long-term outcomes.

Citations

Citations to this article as recorded by  
  • Medical nanoscale materials for virus-induced cervical cancer therapeutic modalities: For targeting delivery
    Adane Adugna, Mamaru Getinet, Gashaw Azanaw Amare, Mohammed Jemal
    OpenNano.2025; 21: 100221.     CrossRef
  • Nano-drug delivery systems based on biodegradable polymers for the therapy of gynecological malignancies
    Keer Jin, Hanxue Zhang, Yuwei Yang, Yan Gao
    International Journal of Polymeric Materials and Polymeric Biomaterials.2024; 73(14): 1262.     CrossRef
  • Dual stimuli-responsive polymeric prodrug consisting of reversible covalent bonded celastrol for tumor targeted delivery
    Jiangtao Su, Meng Rao, Heshuang Dai, Le Cai, Fan Ye, Lu Ye, Yuchen Hu, Ban Chen, Xiaoxia Guo
    Macromolecular Research.2024; 32(2): 173.     CrossRef
  • Designing nanodiscs as versatile platforms for on-demand therapy
    Qianwen Mu, Haolan Deng, Xiaoyu An, Gang Liu, Chao Liu
    Nanoscale.2024; 16(5): 2220.     CrossRef
  • Innovative strategies for effective paclitaxel delivery: Recent developments and prospects
    Sławomir Wileński, Agnieszka Koper, Paulina Śledzińska, Marek Bebyn, Krzysztof Koper
    Journal of Oncology Pharmacy Practice.2024; 30(2): 367.     CrossRef
  • Platelet-derived drug delivery systems: Pioneering treatment for cancer, cardiovascular diseases, infectious diseases, and beyond
    Yalan Zhu, Lingling Xu, Yong Kang, Qinzhen Cheng, Yiling He, Xiaoyuan Ji
    Biomaterials.2024; 306: 122478.     CrossRef
  • Nanoparticle-Based Immunotherapy for Reversing T-Cell Exhaustion
    Fei Li, Yahong Wang, Dandan Chen, Yunjie Du
    International Journal of Molecular Sciences.2024; 25(3): 1396.     CrossRef
  • Delivery of Nucleic Acid Drugs for Tumor Therapy: Opportunities and Challenges
    Dandan Sun, Wenjia Tan, Jianan Zhao, Yafei Tian, Siqi Li, Zhiqi Zhang, Xinzhe Dong, Xiaonan Liu, Na Liu, Ping Jiao, Jie Ma
    Fundamental Research.2024;[Epub]     CrossRef
  • Effect of high-dose polymeric nanoparticle micellar paclitaxel on improved progression-free survival in patients with optimally resected stage III or IV high-grade carcinoma of the ovary: a prospective cohort study with historical controls
    Soo Jin Park, Joo-Hyuk Son, Tae-Wook Kong, Suk-Joon Chang, Hee Seung Kim
    Frontiers in Oncology.2024;[Epub]     CrossRef
  • Co-Assembly of Cancer Drugs with Cyclo-HH Peptides: Insights from Simulations and Experiments
    Anastasia Vlachou, Vijay Bhooshan Kumar, Om Shanker Tiwari, Sigal Rencus-Lazar, Yu Chen, Busra Ozguney, Ehud Gazit, Phanourios Tamamis
    ACS Applied Bio Materials.2024; 7(4): 2309.     CrossRef
  • Biomarkers in Ovarian Cancer: Towards Personalized Medicine
    Carlos López-Portugués, María Montes-Bayón, Paula Díez
    Proteomes.2024; 12(1): 8.     CrossRef
  • Internalization of transferrin-tagged Myxococcus xanthus encapsulins into mesenchymal stem cells
    Anna N. Gabashvili, Natalya A. Alexandrushkina, Elizaveta N. Mochalova, Daria V. Goliusova, Ekaterina N. Sapozhnikova, Pavel I. Makarevich, Petr I. Nikitin
    Experimental Biology and Medicine.2024;[Epub]     CrossRef
  • Functionalized Polymeric Micelles for Targeted Cancer Therapy: Steps from Conceptualization to Clinical Trials
    Ana Serras, Célia Faustino, Lídia Pinheiro
    Pharmaceutics.2024; 16(8): 1047.     CrossRef
  • Polyesters and Polyester Nano- and Microcarriers for Drug Delivery
    Stanislaw Slomkowski, Teresa Basinska, Mariusz Gadzinowski, Damian Mickiewicz
    Polymers.2024; 16(17): 2503.     CrossRef
  • Pharmacokinetic behaviors of soft nanoparticulate formulations of chemotherapeutics
    Mahua Sarkar, Yang Wang, Oscar Ekpenyong, Dong Liang, Huan Xie
    WIREs Nanomedicine and Nanobiotechnology.2023;[Epub]     CrossRef
  • Cancer treatment and toxicity outlook of nanoparticles
    Neetika, Mamta Sharma, Pankaj Thakur, Paras Gaur, Gokana Mohana Rani, Sarvesh Rustagi, Rishi Kumar Talreja, Vishal Chaudhary
    Environmental Research.2023; 237: 116870.     CrossRef
  • Efficacy and Safety of Nanopaclitaxel Formulation for Cancer Treatment: Evidence From Randomized Clinical Trials
    Xiangmin Deng, Xiaoqin Huang, Xiaoyan Dong, Genxiang Mao, Wenmin Xing
    Nanomedicine.2023; 18(10): 833.     CrossRef
  • Development and Perspectives: Multifunctional Nucleic Acid Nanomedicines for Treatment of Gynecological Cancers
    Tetiana Korzun, Abraham S. Moses, Parham Diba, Ariana L. Sattler, Brennan Olson, Olena R. Taratula, Tanja Pejovic, Daniel L. Marks, Oleh Taratula
    Small.2023;[Epub]     CrossRef
  • Nanoprobe-based molecular imaging for tumor stratification
    Xianbin Ma, Mingchuan Mao, Jiaqi He, Chao Liang, Hai-Yan Xie
    Chemical Society Reviews.2023; 52(18): 6447.     CrossRef
  • Magic shotgun over magic bullet for treatment of ovarian cancer via polymeric nanoparticles
    Bakr Ahmed, Anuradha Sharma, Zakiya Usmani, Garima Sharma, Joga Singh, Radhika Yadav, Indu Sharma, Indu Pal Kaur
    Journal of Drug Delivery Science and Technology.2023; 88: 104945.     CrossRef
  • Safety and Tolerability of Weekly Genexol-PM, a Cremophor-Free Polymeric Micelle Formulation of Paclitaxel, with Carboplatin in Gynecologic Cancer: A Phase I Study
    So Hyun Nam, Shin-Wha Lee, Young-Jae Lee, Yong Man Kim
    Cancer Research and Treatment.2023; 55(4): 1346.     CrossRef
  • Effective Cancer Management: Inimitable Role of Phytochemical Based Nano- Formulations
    Aman Upaganlawar, Satish Polshettiwar, Sushil Raut, Amol Tagalpallewar, Vishal Pande
    Current Drug Metabolism.2022; 23(11): 869.     CrossRef
  • Comparison of triblock copolymeric micelles based on α- and ε-poly(L-lysine): a Cornelian choice
    Franck Marquet, Viorica Patrulea, Gerrit Borchard
    Polymer Journal.2022; 54(2): 199.     CrossRef
  • Neurosurgery at the crossroads of immunology and nanotechnology. New reality in the COVID-19 pandemic
    Vladimir A. Ljubimov, Arshia Ramesh, Saya Davani, Moise Danielpour, Joshua J. Breunig, Keith L. Black
    Advanced Drug Delivery Reviews.2022; 181: 114033.     CrossRef
  • Nanocarriers targeting the diseases of the pancreas
    Nurbanu Demirtürk, Erem Bilensoy
    European Journal of Pharmaceutics and Biopharmaceutics.2022; 170: 10.     CrossRef
  • The development and progress of nanomedicine for esophageal cancer diagnosis and treatment
    Xiaokun Li, Lingmin Chen, Siyuan Luan, Jianfeng Zhou, Xin Xiao, Yushang Yang, Chengyi Mao, Pinhao Fang, Longqi Chen, Xiaoxi Zeng, Huile Gao, Yong Yuan
    Seminars in Cancer Biology.2022; 86: 873.     CrossRef
  • Beyond separation: Membranes towards medicine
    Ying Xie, Zhenyu Chu, Wanqin Jin
    Journal of Membrane Science Letters.2022; 2(1): 100020.     CrossRef
  • Modulation of TLR/NF-κB/NLRP Signaling by Bioactive Phytocompounds: A Promising Strategy to Augment Cancer Chemotherapy and Immunotherapy
    Sajad Fakhri, Seyed Zachariah Moradi, Akram Yarmohammadi, Fatemeh Narimani, Carly E. Wallace, Anupam Bishayee
    Frontiers in Oncology.2022;[Epub]     CrossRef
  • Gold Nanorods for Drug and Gene Delivery: An Overview of Recent Advancements
    Atieh Jahangiri-Manesh, Marziyeh Mousazadeh, Shirinsadat Taji, Abbas Bahmani, Atefeh Zarepour, Ali Zarrabi, Esmaeel Sharifi, Mostafa Azimzadeh
    Pharmaceutics.2022; 14(3): 664.     CrossRef
  • Gene Therapy for Malignant and Benign Gynaecological Disorders: A Systematic Review of an Emerging Success Story
    Ekati Drakopoulou, Nicholas P. Anagnou, Kalliopi I. Pappa
    Cancers.2022; 14(13): 3238.     CrossRef
  • Engineering nanosystems to overcome barriers to cancer diagnosis and treatment
    Suhaila O. Alhaj-Suliman, Emad I. Wafa, Aliasger K. Salem
    Advanced Drug Delivery Reviews.2022; 189: 114482.     CrossRef
  • Metabolic Signatures of Surface-Modified Poly(lactic-co-glycolic acid) Nanoparticles in Differentiated THP-1 Cells Derived with Liquid Chromatography-Mass Spectrometry-based Metabolomics
    Mohammad A. Al-natour, Salah Abdelrazig, Amir M. Ghaemmaghami, Cameron Alexander, Dong-Hyun Kim
    ACS Omega.2022; 7(33): 28806.     CrossRef
  • Pathogen-derived peptides in drug targeting and its therapeutic approach
    Seok-Jun Mun, Euni Cho, Jae-Sung Kim, Chul-Su Yang
    Journal of Controlled Release.2022; 350: 716.     CrossRef
  • Application of single and cooperative different delivery systems for the treatment of intervertebral disc degeneration
    Zongtai Liu, Changfeng Fu
    Frontiers in Bioengineering and Biotechnology.2022;[Epub]     CrossRef
  • Engineering the Tumor Immune Microenvironment through Minimally Invasive Interventions
    Koustav Pal, Rahul A. Sheth
    Cancers.2022; 15(1): 196.     CrossRef
  • Nanoparticle delivery systems to combat drug resistance in ovarian cancer
    Emily M. Miller, Timothy M. Samec, Angela A. Alexander-Bryant
    Nanomedicine: Nanotechnology, Biology and Medicine.2021; 31: 102309.     CrossRef
  • Engineering precision nanoparticles for drug delivery
    Michael J. Mitchell, Margaret M. Billingsley, Rebecca M. Haley, Marissa E. Wechsler, Nicholas A. Peppas, Robert Langer
    Nature Reviews Drug Discovery.2021; 20(2): 101.     CrossRef
  • Clinical applications of nanomedicines in lung cancer treatment
    Mohammad Norouzi, Pierre Hardy
    Acta Biomaterialia.2021; 121: 134.     CrossRef
  • Lyophilization stabilizes clinical‐stage core‐crosslinked polymeric micelles to overcome cold chain supply challenges
    Tarun Ojha, Qizhi Hu, Claudio Colombo, Jan Wit, Michiel van Geijn, Mies J. van Steenbergen, Mahsa Bagheri, Hiltrud Königs‐Werner, Eva Miriam Buhl, Ruchi Bansal, Yang Shi, Wim E. Hennink, Gert Storm, Cristianne J. F. Rijcken, Twan Lammers
    Biotechnology Journal.2021;[Epub]     CrossRef
  • Development of next generation nanomedicine-based approaches for the treatment of cancer: we've barely scratched the surface
    Shannon R. Tracey, Peter Smyth, Caroline J. Barelle, Christopher J. Scott
    Biochemical Society Transactions.2021; 49(5): 2253.     CrossRef
  • Impact of Value Frameworks on the Magnitude of Clinical Benefit: Evaluating a Decade of Randomized Trials for Systemic Therapy in Solid Malignancies
    Ellen Cusano, Chelsea Wong, Eddy Taguedong, Marcus Vaska, Tasnima Abedin, Nancy Nixon, Safiya Karim, Patricia Tang, Daniel Y. C. Heng, Doreen Ezeife
    Current Oncology.2021; 28(6): 4894.     CrossRef
  • Engineering Considerations to Produce Bioactive Compounds from Plant Cell Suspension Culture in Bioreactors
    Elizabeth Alejandra Motolinía-Alcántara, Carlos Omar Castillo-Araiza, Mario Rodríguez-Monroy, Angélica Román-Guerrero, Francisco Cruz-Sosa
    Plants.2021; 10(12): 2762.     CrossRef
  • Clinical applications of nanomedicine in cancer therapy
    Mohammad Norouzi, Mehrnaz Amerian, Mahshid Amerian, Fatemeh Atyabi
    Drug Discovery Today.2020; 25(1): 107.     CrossRef
  • Targeting and extending the eukaryotic druggable genome with natural products: cytoskeletal targets of natural products
    April L. Risinger, Lin Du
    Natural Product Reports.2020; 37(5): 634.     CrossRef
  • Improvement of Paclitaxel-Associated Adverse Reactions (ADRs) via the Use of Nano-Based Drug Delivery Systems: A Systematic Review and Network Meta-Analysis


    Pi-Ling Chou, Ya-Ping Huang, Meng-Hsuan Cheng, Kun-Ming Rau, Yi-Ping Fang
    International Journal of Nanomedicine.2020; Volume 15: 1731.     CrossRef
  • Polymeric micelles for the delivery of poorly soluble drugs: From nanoformulation to clinical approval
    Duhyeong Hwang, Jacob D. Ramsey, Alexander V. Kabanov
    Advanced Drug Delivery Reviews.2020; 156: 80.     CrossRef
  • Nanoparticles in precision medicine for ovarian cancer: From chemotherapy to immunotherapy
    Yuan Li, Yan Gao, Xi Zhang, Hongyan Guo, Huile Gao
    International Journal of Pharmaceutics.2020; 591: 119986.     CrossRef
  • Synthesis of PCL–PEG–PCL Triblock Copolymer via Organocatalytic Ring-Opening Polymerization and Its Application as an Injectable Hydrogel—An Interdisciplinary Learning Trial
    Kaiting Wu, Lin Yu, Jiandong Ding
    Journal of Chemical Education.2020; 97(11): 4158.     CrossRef
  • Phytochemical-Based Nanomedicine for Advanced Cancer Theranostics: Perspectives on Clinical Trials to Clinical Use


    Madhusmita Dhupal, Devasish Chowdhury
    International Journal of Nanomedicine.2020; Volume 15: 9125.     CrossRef
  • Synthesis and anti-cancer evaluation of folic acid-peptide- paclitaxel conjugates for addressing drug resistance
    Yuxuan Dai, Xingguang Cai, Xinzhou Bi, Chunxia Liu, Na Yue, Ying Zhu, Jiaqi Zhou, Mian Fu, Wenlong Huang, Hai Qian
    European Journal of Medicinal Chemistry.2019; 171: 104.     CrossRef
  • A Phase II Study of Genexol-PM and Cisplatin as Induction Chemotherapy in Locally Advanced Head and Neck Squamous Cell Carcinoma
    Bhumsuk Keam, Keun-Wook Lee, Se-Hoon Lee, Jin-Soo Kim, Jin Ho Kim, Hong-Gyun Wu, Keun-Yong Eom, Suzy Kim, Soon-Hyun Ahn, Eun-Jae Chung, Seong Keun Kwon, Woo-Jin Jeong, Young Ho Jung, Ji-Won Kim, Dae Seog Heo
    The Oncologist.2019; 24(6): 751.     CrossRef
  • Polymeric Hybrid Nanomicelles for Cancer Theranostics: An Efficient and Precise Anticancer Strategy for the Codelivery of Doxorubicin/miR-34a and Magnetic Resonance Imaging
    Xiaoxue Xie, Yu Chen, Zhongyuan Chen, Yi Feng, Jing Wang, Tingting Li, Shun Li, Xiang Qin, Chunhui Wu, Chuan Zheng, Jie Zhu, Fengming You, Yiyao Liu, Hong Yang
    ACS Applied Materials & Interfaces.2019; 11(47): 43865.     CrossRef
  • Micellar paclitaxel in the treatment of patients with tumors of the female reproductive system
    A. G. Kedrova, S. E. Krasilnikov, D. A. Astakhov, V. V. Kosyy
    Tumors of female reproductive system.2019; 15(3): 37.     CrossRef
  • Improving Drug Delivery of Micellar Paclitaxel against Non‐Small Cell Lung Cancer by Coloading Itraconazole as a Micelle Stabilizer and a Tumor Vascular Manipulator
    Ling Zhang, Zhengsheng Liu, Chao Kong, Chun Liu, Kuan Yang, Huijun Chen, Jinfeng Huang, Feng Qian
    Small.2018;[Epub]     CrossRef
  • 13,129 View
  • 353 Download
  • 57 Web of Science
  • 54 Crossref
Close layer
Feasibility and Efficacy of Eribulin Mesilate in Korean Patients with Metastatic Breast Cancer: Korean Multi-center Phase IV Clinical Study Results
Yeon Hee Park, Tae Yong Kim, Young-Hyuck Im, Keun-Seok Lee, In Hae Park, Joohyuk Sohn, Soo-Hyeon Lee, Seock-Ah Im, Jee Hyun Kim, Se Hyun Kim, Soo Jung Lee, Su-Jin Koh, Ki Hyeong Lee, Yoon Ji Choi, Eun Kyung Cho, Suee Lee, Seok Yun Kang, Jae Hong Seo, Sung-Bae Kim, Kyung Hae Jung
Cancer Res Treat. 2017;49(2):423-429.   Published online August 3, 2016
DOI: https://doi.org/10.4143/crt.2016.191
AbstractAbstract PDFPubReaderePub
Purpose
Eribulin mesilate was approved for the treatment of patients with locally advanced or metastatic breast cancer (MBC),who had received at least two chemotherapeutic regimens, including anthracycline and taxane. On the other hand, the efficacy and safety information of eribulin in Korean patients is limited by the lack of clinical trials.
Materials and Methods
In this multicenter, open-label, single-arm, phase IV study, locally advanced or MBC patients were enrolled between June 2013 and April 2014 from 14 centers in Korea. One point four mg/m2 dose of eribulin was administered on days 1 and 8 of every 21 days. The primary endpoint was the frequency and intensity of the treatment emergent adverse event. The secondary endpoint was the disease control rate, which included the rate of complete responses, partial responses, and stable disease.
Results
A total of 101 patients received at least one dose of eribulin and were included in the safety set. The patients received a total of 543 treatment cycles, with a median of three cycles (range, 1 to 31 cycles). The most common adverse event was neutropenia (91.1% of patients, 48.3% of cycles). The frequent non-hematological adverse events included alopecia, decrease in appetite, fatigue/asthenia, and myalgia/arthralgia. The peripheral neuropathy of any grade occurred in 27 patients (26.7%), including grade 3 in two patients. Disease control rate was 52.7% and 51.3% of patients in the full analysis set and per-protocol set, respectively.
Conclusion
This study demonstrated the feasible safety profile and activity of eribulin in Korean patients with MBC.

Citations

Citations to this article as recorded by  
  • Effectiveness and healthcare costs of eribulin versus capecitabine among metastatic breast cancer patients in Taiwan
    Yu-Ju Lin, Chun-Nan Kuo, Yu Ko
    The Breast.2021; 57: 18.     CrossRef
  • Prognostic and predictive factors of eribulin in patients with heavily pre-treated metastatic breast cancer
    Pei-Hsin Chen, Dah-Cherng Yeh, Heng-Hsin Tung, Chin-Yao Lin
    Medicine.2021; 100(47): e27859.     CrossRef
  • Multifarious targets beyond microtubules—role of eribulin in cancer therapy
    Priya Seshadri, Barnali Deb, Prashant Kumar
    Frontiers in Bioscience-Scholar.2021;[Epub]     CrossRef
  • A nationwide, multicenter retrospective study on the effectiveness and safety of eribulin in Korean breast cancer patients (REMARK)
    Min Ho Park, Soo Jung Lee, Woo Chul Noh, Chang Wan Jeon, Seok Won Lee, Gil Soo Son, Byung-In Moon, Jin Sun Lee, Sung Soo Kang, Young Jin Suh, Geumhee Gwak, Tae Hyun Kim, Young Bum Yoo, Hyun-Ah Kim, Min Young Kim, Ju Yeon Kim, Joon Jeong
    The Breast.2020; 54: 121.     CrossRef
  • Effect of eribulin on patients with metastatic breast cancer: multicenter retrospective observational study in Taiwan
    Kun-Ming Rau, Fu Ou-Yang, Ta-Chung Chao, Yao-Lung Kuo, Tsui-Fen Cheng, Tsu-Yi Chao, Dar-Ren Chen, Yen-Dun Tzeng, Being-Whey Wang, Chun-Yu Liu, Ming-Hung Hu, Yin-Che Lu, Wei-Jen Ou, Chin-Ho Kuo, Chieh-Han Chuang, Jung-Yu Kan, Fang-Ming Chen, Ming-Feng Hou
    Breast Cancer Research and Treatment.2018; 170(3): 583.     CrossRef
  • Incidence and clinical parameters associated with eribulin mesylate-induced peripheral neuropathy
    Bin Zhao, Hong Zhao, Jiaxin Zhao
    Critical Reviews in Oncology/Hematology.2018; 128: 110.     CrossRef
  • Angiomodulators in cancer therapy: New perspectives
    Lenka Varinska, Peter Kubatka, Jan Mojzis, Anthony Zulli, Katarina Gazdikova, Pavol Zubor, Dietrich Büsselberg, Martin Caprnda, Radka Opatrilova, Iveta Gasparova, Martin Klabusay, Martin Pec, Eitan Fibach, Mariusz Adamek, Peter Kruzliak
    Biomedicine & Pharmacotherapy.2017; 89: 578.     CrossRef
  • Eribulin in Advanced Breast Cancer: Safety, Efficacy and New Perspectives
    Ornella Garrone, Emanuela Miraglio, Anna Maria Vandone, Paola Vanella, Daniele Lingua, Marco C Merlano
    Future Oncology.2017; 13(30): 2759.     CrossRef
  • Incidence and relative risk of peripheral neuropathy in cancer patients treated with eribulin: a meta-analysis
    Ling Peng, Yun Hong, Xianghua Ye, Peng Shi, Junyan Zhang, Yina Wang, Qiong Zhao
    Oncotarget.2017; 8(67): 112076.     CrossRef
  • 11,696 View
  • 330 Download
  • 6 Web of Science
  • 9 Crossref
Close layer
Phase II Study of Irinotecan and Cisplatin Combination Chemotherapy in Metastatic, Unresectable Esophageal Cancer
Miso Kim, Bhumsuk Keam, Tae-Min Kim, Hoon-Gu Kim, Jin-Soo Kim, Sung Sook Lee, Seong Hoon Shin, Min Kyoung Kim, Keon Uk Park, Dong-Wan Kim, Hwan Jung Yun, Jong Seok Lee, Dae Seog Heo
Cancer Res Treat. 2017;49(2):416-422.   Published online July 28, 2016
DOI: https://doi.org/10.4143/crt.2016.121
AbstractAbstract PDFPubReaderePub
Purpose
The objective of this multicenter phase II study was to evaluate the efficacy and safety of irinotecan and cisplatin combination chemotherapy in metastatic, unresectable esophageal cancer.
Materials and Methods
Patients were treated with irinotecan 65 mg/m2 and cisplatin 30 mg/m2 on days 1 and 8 of each 21-day treatment cycle. The primary endpoint was response rate, and secondary endpoints were survival, duration of response, initial metabolic response rate, and toxicity.
Results
A total of 27 patients with squamous cell histology were enrolled in the study. The median age of the patients was 61 years. The objective response rate of the 20 patients in the perprotocol group was 30.0% (90% confidence interval [CI], 13.2 to 46.9). The median follow-up duration was 10.0 months, and the median progression-free survival and overall survival were 4.5 months (95% CI, 1.6 to 6.2) and 8.8 months (95% CI, 4.7 to 10.5), respectively. Four of 13 patients (30.8%) evaluated showed initial metabolic response. The median duration of response for partial responders was 5.0 months (range, 3.4 to 8.0 months). The following grade 3/4 treatment-related hematologic toxicities were reported: neutropenia (40.7%), anaemia (22.2%), and thrombocytopenia (7.4%). Two patients experienced febrile neutropenia. The most common grade 3/4 non-hematologic toxicities were asthenia (14.8%) and diarrhoea (11.1%).
Conclusion
Irinotecan and cisplatin combination chemotherapy showed modest anti-tumour activity and manageable toxicity for patients with metastatic, unresectable esophageal cancer.

Citations

Citations to this article as recorded by  
  • Potent molecular-targeted therapies for advanced esophageal squamous cell carcinoma
    Akira Ooki, Hiroki Osumi, Keisho Chin, Masayuki Watanabe, Kensei Yamaguchi
    Therapeutic Advances in Medical Oncology.2023;[Epub]     CrossRef
  • Efficacy and safety of irinotecan combined with raltitrexed or irinotecan monotherapy for salvage chemotherapy of esophageal squamous cell cancer: A prospective, open label, randomized phase II study
    Xichao Dai, Leilei Tao, Jinqiu Wang, Wenjuan Wu, Weigang Bian, Xichun Dai, Surong Chen
    Cancer Medicine.2023; 12(15): 16108.     CrossRef
  • Cisplatin-based combination therapy for cancer
    Minerva, Amrita Bhat, Sonali Verma, Gresh Chander, Rajeshwer Singh Jamwal, Bhawani Sharma, Audesh Bhat, Taruna Katyal, Rakesh Kumar, Ruchi Shah
    Journal of Cancer Research and Therapeutics.2023; 19(3): 530.     CrossRef
  • The development and progress of nanomedicine for esophageal cancer diagnosis and treatment
    Xiaokun Li, Lingmin Chen, Siyuan Luan, Jianfeng Zhou, Xin Xiao, Yushang Yang, Chengyi Mao, Pinhao Fang, Longqi Chen, Xiaoxi Zeng, Huile Gao, Yong Yuan
    Seminars in Cancer Biology.2022; 86: 873.     CrossRef
  • Rh-Endostatin Plus Irinotecan/Cisplatin as Second-Line Therapy for Advanced Esophageal Squamous Cell Carcinoma: An Open-Label, Phase II Study
    Zhihuang Hu, Si Sun, Xinmin Zhao, Hui Yu, Xianghua Wu, Jialei Wang, Jianhua Chang, Huijie Wang
    The Oncologist.2022; 27(4): 253.     CrossRef
  • Study of PD-1 Inhibitors in Combination with Chemoradiotherapy/Chemotherapy in Patients with Esophageal Squamous Carcinoma
    Tianhui Wei, Wenqi Ti, Qingxu Song, Yufeng Cheng
    Current Oncology.2022; 29(5): 2920.     CrossRef
  • Combined treatment with niclosamide and camptothecin enhances anticancer effect in U87 MG human glioblastoma cells
    Laura Valdez, Benxu Cheng, Daniela Gonzalez, Reanna Rodriguez, Paola Campano, Andrew Tsin, Xiaoqian Fang
    Oncotarget.2022; 13(1): 642.     CrossRef
  • Nivolumab for esophageal squamous cell carcinoma and the predictive role of PD-L1 or CD8 expression in its therapeutic effect
    Jiyun Lee, Binnari Kim, Hyun Ae Jung, Yoon La Choi, Jong-Mu Sun
    Cancer Immunology, Immunotherapy.2021; 70(5): 1203.     CrossRef
  • Advances in Our Understanding of the Molecular Mechanisms of Action of Cisplatin in Cancer Therapy
    Paul B Tchounwou, Shaloam Dasari, Felicite K Noubissi, Paresh Ray, Sanjay Kumar
    Journal of Experimental Pharmacology.2021; Volume 13: 303.     CrossRef
  • SHR‐1316, an anti‐PD‐L1 antibody, plus chemotherapy as the first‐line treatment for advanced esophageal squamous cell carcinoma: A multicentre, phase 2 study
    Lan Mu, Yan Song, Kuaile Zhao, Ying Liu, Qingxia Fan, Xi Wang, Qun Li, Xiaopeng Wang, Jing Huang
    Thoracic Cancer.2021; 12(9): 1373.     CrossRef
  • Self-targeted polymersomal co-formulation of doxorubicin, camptothecin and FOXM1 aptamer for efficient treatment of non-small cell lung cancer
    Mahsa Shahriari, Seyed Mohammad Taghdisi, Khalil Abnous, Mohammad Ramezani, Mona Alibolandi
    Journal of Controlled Release.2021; 335: 369.     CrossRef
  • Clinical efficacy of irinotecan plus raltitrexed chemotherapy in refractory esophageal squamous cell cancer
    Min Liu, Qingqing Jia, Xiaolin Wang, Changjiang Sun, Jianqi Yang, Yanliang Chen, Ying Li, Lingfeng Min, Xizhi Zhang, Caiyun Zhu, Johannes Artiaga Gubat, Yong Chen
    Anti-Cancer Drugs.2020; 31(4): 403.     CrossRef
  • Jiawei Xianglian Decoction (JWXLD), a Traditional Chinese Medicine (TCM), Alleviates CPT‐11‐Induced Diarrhea in Mice
    Jinhua Lu, Zechen Lin, Siyu Huang, Yiwei Shen, Jing Jiang, Shengyou Lin, Oliver Micke
    Evidence-Based Complementary and Alternative Medicine.2020;[Epub]     CrossRef
  • Treatment of esophageal cancer with multiple liver metastases: a case experience of sustained complete response
    Jiangfang Wang, Chaoyang Xu
    Journal of International Medical Research.2020;[Epub]     CrossRef
  • Development of chemotherapeutics for unresectable advanced esophageal cancer
    Hiroshi Imazeki, Ken Kato
    Expert Review of Anticancer Therapy.2020; 20(12): 1083.     CrossRef
  • Phase II clinical trial using camrelizumab combined with apatinib and chemotherapy as the first‐line treatment of advanced esophageal squamous cell carcinoma
    Bo Zhang, Ling Qi, Xi Wang, Jianping Xu, Yun Liu, Lan Mu, Xingyuan Wang, Lidan Bai, Jing Huang
    Cancer Communications.2020; 40(12): 711.     CrossRef
  • Systemic treatment of advanced esophageal squamous cell carcinoma: chemotherapy, molecular-targeting therapy and immunotherapy
    Hidekazu Hirano, Ken Kato
    Japanese Journal of Clinical Oncology.2019; 49(5): 412.     CrossRef
  • Carboxylesterase and UDP‐glucuronosyltransferases mediated metabolism of irinotecan: In vitro and in vivo insights from quantitative ultra‐performance liquid chromatography–mass spectrometry analysis
    Yifeng Qin, An Kang, Guisheng Zhou, Huan Wang, Wei Wei, Yujie Cao, Yanyan Chen, Jing Wang, Yajun Shi, Yuping Tang, Jianqin Jiang
    Biomedical Chromatography.2018;[Epub]     CrossRef
  • A combination of irinotecan/cisplatinum and irinotecan/temozolomide or tumor-targeting Salmonella typhimurium A1-R arrest doxorubicin- and temozolomide-resistant myxofibrosarcoma in a PDOX mouse model
    Tasuku Kiyuna, Yasunori Tome, Takashi Murakami, Kentaro Miyake, Kentaro Igarashi, Kei Kawaguchi, Hiromichi Oshiro, Takashi Higuchi, Masuyo Miyake, Norihiko Sugisawa, Zhiying Zhang, Sahar Razmjooei, Sintawat Wangsiricharoen, Bartosz Chmielowski, Scott D. N
    Biochemical and Biophysical Research Communications.2018; 505(3): 733.     CrossRef
  • Human non‑small cell lung cancer cells can be sensitized to camptothecin by modulating autophagy
    Yi-Han Chiu, Shih-Hsien Hsu, Hsiao-Wei Hsu, Kuo-Chin Huang, Wangta Liu, Chang-Yi Wu, Wei-Pang Huang, Jeff Chen, Bing-Hung Chen, Chien-Chih Chiu
    International Journal of Oncology.2018;[Epub]     CrossRef
  • ECRG2 enhances the anti-cancer effects of cisplatin in cisplatin-resistant esophageal cancer cells via upregulation of p53 and downregulation of PNCA
    Xin-Fang Hou, Lin-Ping Xu, Hai-Yan Song, Shuai Li, Chen Wu, Ju-Feng Wang
    World Journal of Gastroenterology.2017; 23(10): 1796.     CrossRef
  • Combination of histoculture drug response assay and qPCR as an effective method to screen biomarkers for personalized chemotherapy in esophageal cancer
    Bin Wei, Jiru Wang, Xiaohui Zhang, Zhaoye Qian, Jingjing Wu, Yuan Sun, Qin Han, Li Wan, Jing Zhu, Yong Gao, Xiaofei Chen
    Oncology Letters.2017;[Epub]     CrossRef
  • 11,760 View
  • 270 Download
  • 27 Web of Science
  • 22 Crossref
Close layer
Induction of Apoptosis in Intestinal Toxicity to a Histone Deacetylase Inhibitor in a Phase I Study with Pelvic Radiotherapy
Erta Kalanxhi, Karianne Risberg, Imon S. Barua, Svein Dueland, Stein Waagene, Solveig Norheim Andersen, Solveig J. Pettersen, Jessica M. Lindvall, Kathrine Røe Redalen, Kjersti Flatmark, Anne Hansen Ree
Cancer Res Treat. 2017;49(2):374-386.   Published online July 28, 2016
DOI: https://doi.org/10.4143/crt.2016.080
AbstractAbstract PDFPubReaderePub
Purpose
When integrating molecularly targeted compounds in radiotherapy, synergistic effects of the systemic agent and radiation may extend the limits of patient tolerance, increasing the demand for understanding the pathophysiological mechanisms of treatment toxicity. In this Pelvic Radiation and Vorinostat (PRAVO) study, we investigated mechanisms of adverse effects in response to the histone deacetylase (HDAC) inhibitor vorinostat (suberoylanilide hydroxamic acid, SAHA) when administered as a potential radiosensitiser.
Materials and Methods
This phase I study for advanced gastrointestinal carcinoma was conducted in sequential patient cohorts exposed to escalating doses of vorinostat combined with standard-fractionated palliative radiotherapy to pelvic target volumes. Gene expression microarray analysis of the study patient peripheral blood mononuclear cells (PBMC) was followed by functional validation in cultured cell lines and mice treated with SAHA.
Results
PBMC transcriptional responses to vorinostat, including induction of apoptosis, were confined to the patient cohort reporting dose-limiting intestinal toxicities. At relevant SAHA concentrations, apoptotic features (annexin V staining and caspase 3/7 activation, but not poly-(ADP-ribose)-polymerase cleavage) were observed in cultured intestinal epithelial cells. Moreover, SAHA-treated mice displayed significant weight loss.
Conclusion
The PRAVO study design implemented a strategy to explore treatment toxicity caused by an HDAC inhibitor when combined with radiotherapy and enabled the identification of apoptosis as a potential mechanism responsible for the dose-limiting effects of vorinostat. To the best of our knowledge, this is the first report deciphering mechanisms of normal tissue adverse effects in response to an HDAC inhibitor within a combined-modality treatment regimen.

Citations

Citations to this article as recorded by  
  • Effect of MicroRNA-210 on the Growth of Ovarian Cancer Cells and the Efficacy of Radiotherapy
    Yinlong Zhao, Shirui Liu, Yu Wen, Lili Zhong
    Gynecologic and Obstetric Investigation.2021; 86(1-2): 71.     CrossRef
  • Valproic Acid Downregulates Cytokine Expression in Human Macrophages Infected with Dengue Virus
    Félix G. Delgado, Paola Cárdenas, Jaime E. Castellanos
    Diseases.2018; 6(3): 59.     CrossRef
  • Synthesis, cytotoxic activity, and mode of action of new Santacruzamate A analogs
    Silmara N. Andrade, Fernanda C. G. Evangelista, Diego Seckler, Deisielly R. Marques, Túlio R. Freitas, Renata R. Nunes, Júlia T. Oliveira, Rosy I. M. A. Ribeiro, Hélio B. Santos, Ralph G. Thomé, Alex G. Taranto, Fabio V. Santos, Gustavo H. R. Viana, Rossi
    Medicinal Chemistry Research.2018; 27(11-12): 2397.     CrossRef
  • Radiosensitization In Vivo by Histone Deacetylase Inhibition with No Increase in Early Normal Tissue Radiation Toxicity
    Blaz Groselj, Jia-Ling Ruan, Helen Scott, Jessica Gorrill, Judith Nicholson, Jacqueline Kelly, Selvakumar Anbalagan, James Thompson, Michael R.L. Stratford, Sarah J. Jevons, Ester M. Hammond, Cheryl L. Scudamore, Martin Kerr, Anne E. Kiltie
    Molecular Cancer Therapeutics.2018; 17(2): 381.     CrossRef
  • 14,985 View
  • 212 Download
  • 4 Web of Science
  • 4 Crossref
Close layer
Phase I Study of OPB-31121, an Oral STAT3 Inhibitor, in Patients with Advanced Solid Tumors
Do-Youn Oh, Se-Hoon Lee, Sae-Won Han, Mi-Jung Kim, Tae-Min Kim, Tae-You Kim, Dae Seog Heo, Miyuki Yuasa, Yasuo Yanagihara, Yung-Jue Bang
Cancer Res Treat. 2015;47(4):607-615.   Published online February 26, 2015
DOI: https://doi.org/10.4143/crt.2014.249
AbstractAbstract PDFPubReaderePub
Purpose
OPB-31121 is an oral STAT3 inhibitor with a good preclinical antitumor activity. This phase I dose-escalation study of OPB-31121 was conducted to determine maximum-tolerated dose (MTD), safety, pharmacokinetics, and preliminary antitumor efficacy in patients with advanced solid tumors. Materials and Methods Patients received OPB-31121 once daily for 28 days of each cycle followed by 2 weeks rest. A standard 3+3 design was used for dose-escalation. Safety and response were evaluated by the National Cancer Institute–Common Terminology Criteria for Adverse Events (NCICTCAE) ver. 3.0 and Response Evaluation Criteria in Solid Tumor (RECIST) ver. 1.0, respectively.
Results
Twenty-five patients were treated with OPB-31121 at five dose levels: 100 mg (n=4), 200 mg (n=3), 400 mg (n=3), 600 mg (n=7), and 800 mg (n=8). Seven patients discontinued treatment during cycle 1 for various reasons other than study drug-related adverse events. Among 18 patients who were evaluable for dose-limiting toxicity (DLT), three DLTs were observed: one DLT (grade 3 vomiting) at 600 mg and two DLTs (grade 3 vomiting, grade 3 diarrhea) at 800 mg. The MTD was determined as 800 mg/day. Common adverse events were gastrointestinal adverse event including nausea (84%), vomiting (80%), and diarrhea (72%). Pharmacokinetics did not demonstrate dose-proportionality of OPB-31121. Eight patients had stable disease and 10 patients had disease progression. Two patients (1 colon cancer, 1 rectal cancer) showed tumor shrinkage. One gastric cancer patient continued treatment up to cycle 13 before disease progression. Conclusion This study demonstrates feasibility of STAT3 inhibition in patients with advanced solid tumor. OPB-31121, at the MTD of 800 mg/day, was safe and relatively well tolerated, and has a preliminary antitumor activity.

Citations

Citations to this article as recorded by  
  • Unraveling the complexity of STAT3 in cancer: molecular understanding and drug discovery
    Yamei Hu, Zigang Dong, Kangdong Liu
    Journal of Experimental & Clinical Cancer Research.2024;[Epub]     CrossRef
  • Hepatocellular Carcinoma: Old and Emerging Therapeutic Targets
    Greta Pessino, Claudia Scotti, Maristella Maggi
    Cancers.2024; 16(5): 901.     CrossRef
  • Impact of STAT-signaling pathway on cancer-associated fibroblasts in colorectal cancer and its role in immunosuppression
    Damián Sánchez-Ramírez, Mónica G Mendoza-Rodríguez, Omar R Alemán, Fernando A Candanedo-González, Miriam Rodríguez-Sosa, Juan José Montesinos-Montesinos, Mauricio Salcedo, Ismael Brito-Toledo, Felipe Vaca-Paniagua, Luis I Terrazas
    World Journal of Gastrointestinal Oncology.2024; 16(5): 1705.     CrossRef
  • Targeted Treatment against Cancer Stem Cells in Colorectal Cancer
    Julia Martínez-Pérez, Carlos Torrado, María A. Domínguez-Cejudo, Manuel Valladares-Ayerbes
    International Journal of Molecular Sciences.2024; 25(11): 6220.     CrossRef
  • Interleukin-6 serves as a critical factor in various cancer progression and therapy
    Asma’a H. Mohamed, Abdulrahman T. Ahmed, Waleed Al Abdulmonem, Dmitry Olegovich Bokov, Alaa Shafie, Hussein Riyadh Abdul Kareem Al-Hetty, Chou-Yi Hsu, Mohammed Alissa, Shahid Nazir, Mohammad Chand Jamali, Mustafa Mudhafar
    Medical Oncology.2024;[Epub]     CrossRef
  • Targeting cytokine and chemokine signaling pathways for cancer therapy
    Ming Yi, Tianye Li, Mengke Niu, Haoxiang Zhang, Yuze Wu, Kongming Wu, Zhijun Dai
    Signal Transduction and Targeted Therapy.2024;[Epub]     CrossRef
  • Recent advances in targeted drug delivery systems for multiple myeloma
    Ashruti Pant, Aayushi Laliwala, Sarah A. Holstein, Aaron M. Mohs
    Journal of Controlled Release.2024; 376: 215.     CrossRef
  • A strategic review of STAT3 signaling inhibition by phytochemicals for cancer prevention and treatment: Advances and insights
    Suryaa Manoharan, Ekambaram Perumal
    Fitoterapia.2024; 179: 106265.     CrossRef
  • Expression of STAT3, IL27p28 and IL12p35 is deregulated and linked to autoimmune markers in chronic spontaneous urticaria
    Sahar Rastgoo, Mojgan Mohammadi, Marcus Maurer, Mahdi Atabaki, Jalil Tavakkol-Afshari, Maryam Khoshkhui
    Clinical and Experimental Dermatology.2024;[Epub]     CrossRef
  • The STAT family: Key transcription factors mediating crosstalk between cancer stem cells and tumor immune microenvironment
    Mengxuan Zhu, Suyao Li, Xin Cao, Khalid Rashid, Tianshu Liu
    Seminars in Cancer Biology.2023; 88: 18.     CrossRef
  • Dual inhibition of MYC and SLC39A10 by a novel natural product STAT3 inhibitor derived from Chaetomium globosum suppresses tumor growth and metastasis in gastric cancer
    Xiaoqing Guan, Jing Yang, Weiyi Wang, Bing Zhao, Shiyu Hu, Dehua Yu, Li Yuan, Yunfu Shi, Jingli Xu, Jinyun Dong, Jinxin Wang, Xiang-Dong Cheng, Jiang-Jiang Qin
    Pharmacological Research.2023; 189: 106703.     CrossRef
  • JAK/STAT pathway: Extracellular signals, diseases, immunity, and therapeutic regimens
    Qian Hu, Qihui Bian, Dingchao Rong, Leiyun Wang, Jianan Song, Hsuan-Shun Huang, Jun Zeng, Jie Mei, Peng-Yuan Wang
    Frontiers in Bioengineering and Biotechnology.2023;[Epub]     CrossRef
  • Brevilin A is a potent anti-metastatic CRC agent that targets the VEGF-IL6-STAT3 axis in the HSCs-CRC interplay
    Xueying Fan, Mingjing Meng, Baoting Li, Hui Chen, Jincheng Tan, Keyang Xu, Shilin Xiao, Hiu-Yee Kwan, Zhongqiu Liu, Tao Su
    Journal of Translational Medicine.2023;[Epub]     CrossRef
  • Application of Nano-Antibodies for Cancer Immunotherapy
    Sunanda Singh, Samara P. Singh, Ashutosh S. Parihar
    Current Tissue Microenvironment Reports.2023; 4(2): 17.     CrossRef
  • Exploring the dynamic interplay between cancer stem cells and the tumor microenvironment: implications for novel therapeutic strategies
    Yan-Ruide Li, Ying Fang, Zibai Lyu, Yichen Zhu, Lili Yang
    Journal of Translational Medicine.2023;[Epub]     CrossRef
  • C188-9, a specific inhibitor of STAT3 signaling, prevents thermal burn-induced skeletal muscle wasting in mice
    Yuko Ono, Masafumi Saito, Kazuho Sakamoto, Yuko Maejima, Shingen Misaka, Kenju Shimomura, Nobuto Nakanishi, Shigeaki Inoue, Joji Kotani
    Frontiers in Pharmacology.2022;[Epub]     CrossRef
  • Signaling Pathways and Targeted Therapies for Stem Cells in Prostate Cancer
    Madhuvanthi Giridharan, Vasu Rupani, Satarupa Banerjee
    ACS Pharmacology & Translational Science.2022; 5(4): 193.     CrossRef
  • Insights into the role of STAT3 in intrahepatic cholangiocarcinoma (Review)
    Ranzhiqiang Yang, Yinghui Song, Kashif Shakoor, Weimin Yi, Chuang Peng, Sulai Liu
    Molecular Medicine Reports.2022;[Epub]     CrossRef
  • Inhibition of STAT3-ferroptosis negative regulatory axis suppresses tumor growth and alleviates chemoresistance in gastric cancer
    Shumin Ouyang, Huaxuan Li, Linlin Lou, Qiuyao Huang, Zhenhua Zhang, Jianshan Mo, Min Li, Jiaye Lu, Kai Zhu, Yunjie Chu, Wen Ding, Jianzheng Zhu, Ziyou Lin, Lin Zhong, Junjian Wang, Peibin Yue, James Turkson, Peiqing Liu, Yuanxiang Wang, Xiaolei Zhang
    Redox Biology.2022; 52: 102317.     CrossRef
  • Digesting the Role of JAK-STAT and Cytokine Signaling in Oral and Gastric Cancers
    Yanhong Ni, Jun T. Low, John Silke, Lorraine A. O’Reilly
    Frontiers in Immunology.2022;[Epub]     CrossRef
  • A Broad-Based Characterization of a Cell-Penetrating, Single Domain Camelid Bi-Specific Antibody Monomer That Targets STAT3 and KRAS Dependent Cancers
    Sunanda Singh, Genoveva Murillo, Justin Richner, Samara P. Singh, Erica Berleth, Vijay Kumar, Rajendra Mehta, Vijay Ramiya, Ashutosh S. Parihar
    International Journal of Molecular Sciences.2022; 23(14): 7565.     CrossRef
  • Molecular mechanisms underlying the action of carcinogens in gastric cancer with a glimpse into targeted therapy
    Elham Patrad, Solmaz Khalighfard, Taghi Amiriani, Vahid Khori, Ali Mohammad Alizadeh
    Cellular Oncology.2022; 45(6): 1073.     CrossRef
  • STAT3 and Its Targeting Inhibitors in Oral Squamous Cell Carcinoma
    Mingjing Jiang, Bo Li
    Cells.2022; 11(19): 3131.     CrossRef
  • The Role of IL-6 in Cancer Cell Invasiveness and Metastasis—Overview and Therapeutic Opportunities
    Magdalena Rašková, Lukáš Lacina, Zdeněk Kejík, Anna Venhauerová, Markéta Skaličková, Michal Kolář, Milan Jakubek, Daniel Rosel, Karel Smetana, Jan Brábek
    Cells.2022; 11(22): 3698.     CrossRef
  • An update on investigational therapies that target STAT3 for the treatment of cancer
    Matteo Santoni, Francesca Miccini, Alessia Cimadamore, Francesco Piva, Francesco Massari, Liang Cheng, Antonio Lopez-Beltran, Rodolfo Montironi, Nicola Battelli
    Expert Opinion on Investigational Drugs.2021; 30(3): 245.     CrossRef
  • RETRACTED: GATA4 Regulates Inflammation-Driven Pancreatic Ductal Adenocarcinoma Progression
    Weiliang Jiang, Congying Chen, Li Huang, Jie Shen, Lijuan Yang
    Frontiers in Cell and Developmental Biology.2021;[Epub]     CrossRef
  • Phytochemicals Targeting JAK–STAT Pathways in Inflammatory Bowel Disease: Insights from Animal Models
    Sun Young Moon, Kwang Dong Kim, Jiyun Yoo, Jeong-Hyung Lee, Cheol Hwangbo
    Molecules.2021; 26(9): 2824.     CrossRef
  • Development and Validation of an IL6/JAK/STAT3-Related Gene Signature to Predict Overall Survival in Clear Cell Renal Cell Carcinoma
    Chuanchuan Zhan, Chao Xu, Jiajun Chen, Chong Shen, Jinkun Li, Zichu Wang, Xiangrong Ying, Zhengang Luo, Yu Ren, Gangfeng Wu, Haojie Zhang, Manfei Qian
    Frontiers in Cell and Developmental Biology.2021;[Epub]     CrossRef
  • Emerging role of signal transducer and activator of transcription 3 (STAT3) in pituitary adenomas
    Cyndy Liu, Tae Nakano-Tateno, Motoyasu Satou, Constance Chik, Toru Tateno
    Endocrine Journal.2021; 68(10): 1143.     CrossRef
  • The JAK/STAT signaling pathway: from bench to clinic
    Xiaoyi Hu, Jing li, Maorong Fu, Xia Zhao, Wei Wang
    Signal Transduction and Targeted Therapy.2021;[Epub]     CrossRef
  • Interleukin-6 and colorectal cancer development
    I.А. Hromakova, P.P. Sorochan, N.E. Prokhach, I.S. Hromakova
    Український радіологічний та онкологічний журнал.2021; 29(4): 89.     CrossRef
  • Ailanthone suppresses the activity of human colorectal cancer cells through the STAT3 signaling pathway
    Haixiang Ding, Xiuchong Yu, Zhilong Yan
    International Journal of Molecular Medicine.2021;[Epub]     CrossRef
  • Radiation induces an inflammatory response that results in STAT3-dependent changes in cellular plasticity and radioresistance of breast cancer stem-like cells
    Kimberly M. Arnold, Lynn M. Opdenaker, Nicole J. Flynn, Daniel Kwesi Appeah, Jennifer Sims-Mourtada
    International Journal of Radiation Biology.2020; 96(4): 434.     CrossRef
  • Quinazoline Ligands Induce Cancer Cell Death through Selective STAT3 Inhibition and G-Quadruplex Stabilization
    Jan Jamroskovic, Mara Doimo, Karam Chand, Ikenna Obi, Rajendra Kumar, Kristoffer Brännström, Mattias Hedenström, Rabindra Nath Das, Almaz Akhunzianov, Marco Deiana, Kazutoshi Kasho, Sebastian Sulis Sato, Parham L. Pourbozorgi, James E. Mason, Paolo Medini
    Journal of the American Chemical Society.2020; 142(6): 2876.     CrossRef
  • Phosphotyrosine isosteres: past, present and future
    Robert A. Cerulli, Joshua A. Kritzer
    Organic & Biomolecular Chemistry.2020; 18(4): 583.     CrossRef
  • Anticancer activity of dietary xanthone α-mangostin against hepatocellular carcinoma by inhibition of STAT3 signaling via stabilization of SHP1
    Hai Zhang, Yu-ping Tan, Lin Zhao, Lun Wang, Nai-jie Fu, Song-ping Zheng, Xiao-fei Shen
    Cell Death & Disease.2020;[Epub]     CrossRef
  • JAK/STAT signaling in hepatocellular carcinoma
    Justin Jit Hin Tang, Dexter Kai Hao Thng, Jhin Jieh Lim, Tan Boon Toh
    Hepatic Oncology.2020;[Epub]     CrossRef
  • JAK–STAT pathway targeting for the treatment of inflammatory bowel disease
    Azucena Salas, Cristian Hernandez-Rocha, Marjolijn Duijvestein, William Faubion, Dermot McGovern, Severine Vermeire, Stefania Vetrano, Niels Vande Casteele
    Nature Reviews Gastroenterology & Hepatology.2020; 17(6): 323.     CrossRef
  • Targeting STAT3 in cancer and autoimmune diseases
    Tohid Gharibi, Zohreh Babaloo, Arezoo Hosseini, Meghdad Abdollahpour-alitappeh, Vida Hashemi, Faroogh Marofi, Kazem Nejati, Behzad Baradaran
    European Journal of Pharmacology.2020; 878: 173107.     CrossRef
  • Betacellulin drives therapy resistance in glioblastoma
    Qiwen Fan, Zhenyi An, Robyn A Wong, Xujun Luo, Edbert D Lu, Albert Baldwin, Manasi K Mayekar, Franziska Haderk, Kevan M Shokat, Trever G Bivona, William A Weiss
    Neuro-Oncology.2020; 22(4): 457.     CrossRef
  • A novel small molecule STAT3 inhibitor SLSI-1216 suppresses proliferation and tumor growth of triple-negative breast cancer cells through apoptotic induction
    Soo Kyung Park, Woong Sub Byun, Seungbeom Lee, Young Taek Han, Yoo-Seong Jeong, Kyungkuk Jang, Suk-Jae Chung, Jeeyeon Lee, Young-Ger Suh, Sang Kook Lee
    Biochemical Pharmacology.2020; 178: 114053.     CrossRef
  • STAT3 Pathway in Gastric Cancer: Signaling, Therapeutic Targeting and Future Prospects
    Milad Ashrafizadeh, Ali Zarrabi, Sima Orouei, Vahideh Zarrin, Ebrahim Rahmani Moghadam, Amirhossein Zabolian, Shima Mohammadi, Kiavash Hushmandi, Yashar Gharehaghajlou, Pooyan Makvandi, Masoud Najafi, Reza Mohammadinejad
    Biology.2020; 9(6): 126.     CrossRef
  • Towards the Inhibition of Protein–Protein Interactions (PPIs) in STAT3: Insights into a New Class of Benzothiadiazole Derivatives
    Matteo Mori, Ettore Gilardoni, Luca Regazzoni, Alessandro Pedretti, Diego Colombo, Gary Parkinson, Akira Asai, Fiorella Meneghetti, Stefania Villa, Arianna Gelain
    Molecules.2020; 25(15): 3509.     CrossRef
  • STAT3, the Challenge for Chemotherapeutic and Radiotherapeutic Efficacy
    Ping-Lian Yang, Lu-Xin Liu, En-Min Li, Li-Yan Xu
    Cancers.2020; 12(9): 2459.     CrossRef
  • Progress in Understanding the IL-6/STAT3 Pathway in Colorectal Cancer


    Yan Lin, Ziqin He, Jiazhou Ye, Ziyu Liu, Xiaomin She, Xing Gao, Rong Liang
    OncoTargets and Therapy.2020; Volume 13: 13023.     CrossRef
  • Systemic Therapy for Hepatocellular Carcinoma: Advances and Hopes
    Chen-Hao Zhang, Ming Li, You-Pei Lin, Qiang Gao
    Current Gene Therapy.2020; 20(2): 84.     CrossRef
  • Decoration of Anti-CD38 on Nanoparticles Carrying a STAT3 Inhibitor Can Improve the Therapeutic Efficacy Against Myeloma
    Yung-Hsing Huang, Mohammad Reza Vakili, Ommoleila Molavi, Yuen Morrissey, Chengsheng Wu, Igor Paiva, Amir Hasan Soleimani, Forugh Sanaee, Afsaneh Lavasanifar, Raymond Lai
    Cancers.2019; 11(2): 248.     CrossRef
  • Phase I Dose-Finding Study of OPB-111077, a Novel STAT3 Inhibitor, in Patients with Advanced Hepatocellular Carcinoma
    Changhoon Yoo, Jihoon Kang, Ho Yeong Lim, Jee Hyun Kim, Myung-Ah Lee, Kyung-Hun Lee, Tae-You Kim, Baek-Yeol Ryoo
    Cancer Research and Treatment.2019; 51(2): 510.     CrossRef
  • IL-6 signaling contributes to radioresistance of prostate cancer through key DNA repair-associated molecules ATM, ATR, and BRCA 1/2
    Xiaodong Chen, Feng Chen, Yu Ren, Guobin Weng, Lijun Xu, Xiang Xue, Peter C. Keng, Soo Ok Lee, Yuhchyau Chen
    Journal of Cancer Research and Clinical Oncology.2019; 145(6): 1471.     CrossRef
  • Proton pump inhibitor: The dual role in gastric cancer
    Moon Kyung Joo, Jong-Jae Park, Hoon Jai Chun
    World Journal of Gastroenterology.2019; 25(17): 2058.     CrossRef
  • Transcriptional Reprogramming and Novel Therapeutic Approaches for Targeting Prostate Cancer Stem Cells
    Gianluca Civenni, Domenico Albino, Dheeraj Shinde, Ramiro Vázquez, Jessica Merulla, Aleksandra Kokanovic, Sarah N. Mapelli, Giuseppina M. Carbone, Carlo V. Catapano
    Frontiers in Oncology.2019;[Epub]     CrossRef
  • STAT3 as a potential therapeutic target in triple negative breast cancer: a systematic review
    Jiang-Jiang Qin, Li Yan, Jia Zhang, Wei-Dong Zhang
    Journal of Experimental & Clinical Cancer Research.2019;[Epub]     CrossRef
  • JAK/STAT inhibition in macrophages promotes therapeutic resistance by inducing expression of protumorigenic factors
    Emily A. Irey, Chelsea M. Lassiter, Nicholas J. Brady, Pavlina Chuntova, Ying Wang, Todd P. Knutson, Christine Henzler, Thomas S. Chaffee, Rachel I. Vogel, Andrew C. Nelson, Michael A. Farrar, Kathryn L. Schwertfeger
    Proceedings of the National Academy of Sciences.2019; 116(25): 12442.     CrossRef
  • Elevated HOX gene expression in acute myeloid leukemia is associated with NPM1 mutations and poor survival
    Ádám Nagy, Ágnes Ősz, Jan Budczies, Szilvia Krizsán, Gergely Szombath, Judit Demeter, Csaba Bödör, Balázs Győrffy
    Journal of Advanced Research.2019; 20: 105.     CrossRef
  • Antitumor activity of novel pyrazole-based small molecular inhibitors of the STAT3 pathway in patient derived high grade glioma cells
    Liang Zhang, Timothy E. Peterson, Victor M. Lu, Ian F. Parney, David J. Daniels, Ilya Ulasov
    PLOS ONE.2019; 14(7): e0220569.     CrossRef
  • Inhibition of Stat3 Signaling Pathway by Natural Product Pectolinarigenin Attenuates Breast Cancer Metastasis
    Yali Li, Cailing Gan, Yange Zhang, Yan Yu, Chen Fan, Yuanle Deng, Qianyu Zhang, Xi Yu, Yiwen Zhang, Liqun Wang, Fang He, Yongmei Xie, Tinghong Ye, Wenya Yin
    Frontiers in Pharmacology.2019;[Epub]     CrossRef
  • Impact of combination therapy with anti-PD-1 blockade and a STAT3 inhibitor on the tumor-infiltrating lymphocyte status
    Tadashi Ashizawa, Akira Iizuka, Chie Maeda, Emiko Tanaka, Ryota Kondou, Haruo Miyata, Takashi Sugino, Takuya Kawata, Shoichi Deguchi, Koichi Mitsuya, Nakamasa Hayashi, Akira Asai, Mamoru Ito, Ken Yamaguchi, Yasuto Akiyama
    Immunology Letters.2019; 216: 43.     CrossRef
  • Signal Transducer and Activator of Transcription Protein 3 (STAT3): An Update on its Direct Inhibitors as Promising Anticancer Agents
    Arianna Gelain, Matteo Mori, Fiorella Meneghetti, Stefania Villa
    Current Medicinal Chemistry.2019; 26(27): 5165.     CrossRef
  • The effects of signal transducer and activator of transcription three mutations on human platelets
    Floor E. Aleva, Frank L. van de Veerdonk, Yang Li, Rahajeng N. Tunjungputri, Sami Simons, Philip G. De Groot, Mihai M. Netea, Yvonne F. Heijdra, Quirijn de Mast, André J.A.M. van der Ven
    Platelets.2018; 29(6): 602.     CrossRef
  • Mechanisms Linking Obesity and Thyroid Cancer Development and Progression in Mouse Models
    Won Gu Kim, Sheue-yann Cheng
    Hormones and Cancer.2018; 9(2): 108.     CrossRef
  • Targeting the IL-6/JAK/STAT3 signalling axis in cancer
    Daniel E. Johnson, Rachel A. O'Keefe, Jennifer R. Grandis
    Nature Reviews Clinical Oncology.2018; 15(4): 234.     CrossRef
  • Arsenic trioxide attenuates STAT-3 activity and epithelial-mesenchymal transition through induction of SHP-1 in gastric cancer cells
    Sung Ho Kim, Hyo Soon Yoo, Moon Kyung Joo, Taehyun Kim, Jong-Jae Park, Beom Jae Lee, Hoon Jai Chun, Sang Woo Lee, Young-Tae Bak
    BMC Cancer.2018;[Epub]     CrossRef
  • Linker Variation and Structure–Activity Relationship Analyses of Carboxylic Acid-based Small Molecule STAT3 Inhibitors
    Francisco Lopez-Tapia, Christine Brotherton-Pleiss, Peibin Yue, Heide Murakami, Ana Carolina Costa Araujo, Bruna Reis dos Santos, Erin Ichinotsubo, Anna Rabkin, Raj Shah, Megan Lantz, Suzie Chen, Marcus A. Tius, James Turkson
    ACS Medicinal Chemistry Letters.2018; 9(3): 250.     CrossRef
  • Decoy-Based, Targeted Inhibition of STAT3: A New Step forward for B Cell Lymphoma Immunotherapy
    Mario M. Soldevilla, Fernando Pastor
    Molecular Therapy.2018; 26(3): 675.     CrossRef
  • Two decades of research in discovery of anticancer drugs targeting STAT3, how close are we?
    Jenny D. Beebe, Jing-Yuan Liu, Jian-Ting Zhang
    Pharmacology & Therapeutics.2018; 191: 74.     CrossRef
  • Translational and clinical advances in JAK-STAT biology: The present and future of jakinibs
    Massimo Gadina, Catrina Johnson, Daniella Schwartz, Michael Bonelli, Sarfaraz Hasni, Yuka Kanno, Paul Changelian, Arian Laurence, John J O’Shea
    Journal of Leukocyte Biology.2018; 104(3): 499.     CrossRef
  • Targeting JAK-STAT signal transduction in IBD
    Christoffer Soendergaard, Fredrik Holmberg Bergenheim, Jakob Tveiten Bjerrum, Ole Haagen Nielsen
    Pharmacology & Therapeutics.2018; 192: 100.     CrossRef
  • “Do We Know Jack” About JAK? A Closer Look at JAK/STAT Signaling Pathway
    Emira Bousoik, Hamidreza Montazeri Aliabadi
    Frontiers in Oncology.2018;[Epub]     CrossRef
  • Plasticity of Type I Interferon-Mediated Responses in Cancer Therapy: From Anti-tumor Immunity to Resistance
    Megha Budhwani, Roberta Mazzieri, Riccardo Dolcetti
    Frontiers in Oncology.2018;[Epub]     CrossRef
  • STAT3 in Breast Cancer Onset and Progression: A Matter of Time and Context
    Ilenia Segatto, Gustavo Baldassarre, Barbara Belletti
    International Journal of Molecular Sciences.2018; 19(9): 2818.     CrossRef
  • Suppression of STAT3 signaling promotes cellular reprogramming into insulin-producing cells induced by defined transcription factors
    Masaki Miura, Takeshi Miyatsuka, Takehiro Katahira, Shugo Sasaki, Luka Suzuki, Miwa Himuro, Yuya Nishida, Yoshio Fujitani, Taka-aki Matsuoka, Hirotaka Watada
    EBioMedicine.2018; 36: 358.     CrossRef
  • Aptamer-iRNAs as Therapeutics for Cancer Treatment
    Mario M. Soldevilla, Daniel Meraviglia-Crivelli de Caso, Ashwathi P. Menon, Fernando Pastor
    Pharmaceuticals.2018; 11(4): 108.     CrossRef
  • A First-in-Human Phase I Study of OPB-111077, a Small-Molecule STAT3 and Oxidative Phosphorylation Inhibitor, in Patients with Advanced Cancers
    Anthony Tolcher, Keith Flaherty, Geoffrey I. Shapiro, Jordan Berlin, Thomas Witzig, Thomas Habermann, Andrea Bullock, Edwin Rock, Agnes Elekes, Chester Lin, Dusan Kostic, Naoto Ohi, Drew Rasco, Kyriakos P. Papadopoulos, Amita Patnaik, Lon Smith, Gregory M
    The Oncologist.2018; 23(6): 658.     CrossRef
  • Impact of STAT3 phosphorylation in glioblastoma stem cells radiosensitization and patient outcome
    Konstantin Masliantsev, Baptiste Pinel, Anaïs Balbous, Pierre-Olivier Guichet, Gaëlle Tachon, Serge Milin, Julie Godet, Mathilde Duchesne, Antoine Berger, Christos Petropoulos, Michel Wager, Lucie Karayan-Tapon
    Oncotarget.2018; 9(3): 3968.     CrossRef
  • Discovery of an Orally Selective Inhibitor of Signal Transducer and Activator of Transcription 3 Using Advanced Multiple Ligand Simultaneous Docking
    Wenying Yu, Chenglong Li, Wenda Zhang, Yuanzheng Xia, Shanshan Li, Jia-yuh Lin, Keqin Yu, Mu Liu, Lei Yang, Jianguang Luo, Yijun Chen, Hongbin Sun, Lingyi Kong
    Journal of Medicinal Chemistry.2017; 60(7): 2718.     CrossRef
  • Exploiting the Ref-1-APE1 node in cancer signaling and other diseases: from bench to clinic
    Fenil Shah, Derek Logsdon, Richard A. Messmann, Jill C. Fehrenbacher, Melissa L. Fishel, Mark R. Kelley
    npj Precision Oncology.2017;[Epub]     CrossRef
  • Mitochondrial dysfunction induced by a SH2 domain-targeting STAT3 inhibitor leads to metabolic synthetic lethality in cancer cells
    Davide Genini, Lara Brambilla, Erik Laurini, Jessica Merulla, Gianluca Civenni, Shusil Pandit, Rocco D'Antuono, Laurent Perez, David E. Levy, Sabrina Pricl, Giuseppina M. Carbone, Carlo V. Catapano
    Proceedings of the National Academy of Sciences.2017;[Epub]     CrossRef
  • Identification of novel small molecules that inhibit STAT3-dependent transcription and function
    Iryna Kolosenko, Yasmin Yu, Sander Busker, Matheus Dyczynski, Jianping Liu, Martin Haraldsson, Caroline Palm Apergi, Thomas Helleday, Katja Pokrovskaja Tamm, Brent D. G. Page, Dan Grander, Aamir Ahmad
    PLOS ONE.2017; 12(6): e0178844.     CrossRef
  • Antineoplastic effects of CPPTL via the ROS/JNK pathway in acute myeloid leukemia
    Hui-Er Gao, Yue Sun, Ya-Hui Ding, Jing Long, Xiao-Lei Liu, Ming Yang, Qing Ji, Ying-Hui Li, Yue Chen, Quan Zhang, Ying-Dai Gao
    Oncotarget.2017; 8(24): 38990.     CrossRef
  • Stattic and metformin inhibit brain tumor initiating cells by reducing STAT3-phosphorylation
    Verena Leidgens, Judith Proske, Lisa Rauer, Sylvia Moeckel, Kathrin Renner, Ulrich Bogdahn, Markus J. Riemenschneider, Martin Proescholdt, Arabel Vollmann-Zwerenz, Peter Hau, Corinna Seliger
    Oncotarget.2017; 8(5): 8250.     CrossRef
  • Identification of antipsychotic drug fluspirilene as a potential anti-glioma stem cell drug
    Yu Dong, Takuya Furuta, Hemragul Sabit, Tomohiro Kitabayashi, Shabierjiang Jiapaer, Masahiko Kobayashi, Yasushi Ino, Tomoki Todo, Lei Teng, Atsushi Hirao, Shi-Guang Zhao, Mitsutoshi Nakada
    Oncotarget.2017; 8(67): 111728.     CrossRef
  • Feedback Activation of STAT3 as a Cancer Drug-Resistance Mechanism
    Chengguang Zhao, Huameng Li, Huey-Jen Lin, Shulin Yang, Jiayuh Lin, Guang Liang
    Trends in Pharmacological Sciences.2016; 37(1): 47.     CrossRef
  • Targeting transcription factor STAT3 for cancer prevention and therapy
    Edna Zhi Pei Chai, Muthu K. Shanmugam, Frank Arfuso, Arunasalam Dharmarajan, Chao Wang, Alan Prem Kumar, Ramar Perumal Samy, Lina H.K. Lim, Lingzhi Wang, Boon Cher Goh, Kwang Seok Ahn, Kam Man Hui, Gautam Sethi
    Pharmacology & Therapeutics.2016; 162: 86.     CrossRef
  • Molecular Bases for Combinatorial Treatment Strategies in Patients with KRAS Mutant Lung Adenocarcinoma and Squamous Cell Lung Carcinoma
    Chiara Lazzari, Alberto Verlicchi, Anastasios Gkountakos, Sara Pilotto, Mariacarmela Santarpia, Imane Chaib, Jose Luis Ramirez Serrano, Santiago Viteri, Daniela Morales-Espinosa, Claudio Dazzi, Filippo de Marinis, Peng Cao, Niki Karachaliou, Rafael Rosell
    Pulmonary Therapy.2016; 2(1): 1.     CrossRef
  • Preclinical Characterization of 3β-(N-Acetyl l-cysteine methyl ester)-2aβ,3-dihydrogaliellalactone (GPA512), a Prodrug of a Direct STAT3 Inhibitor for the Treatment of Prostate Cancer
    Zilma Escobar, Anders Bjartell, Giacomo Canesin, Susan Evans-Axelsson, Olov Sterner, Rebecka Hellsten, Martin H Johansson
    Journal of Medicinal Chemistry.2016; 59(10): 4551.     CrossRef
  • A Positive TGF-β/c-KIT Feedback Loop Drives Tumor Progression in Advanced Primary Liver Cancer
    Andres Rojas, Pingyu Zhang, Ying Wang, Wai Chin Foo, Nina M. Muñoz, Lianchun Xiao, Jing Wang, Gregory J. Gores, Mien-Chie Hung, Boris Blechacz
    Neoplasia.2016; 18(6): 371.     CrossRef
  • Novel STAT 3 inhibitors for treating gastric cancer
    Catherine Cafferkey, Ian Chau
    Expert Opinion on Investigational Drugs.2016; 25(9): 1023.     CrossRef
  • ‘Acute myeloid leukemia: a comprehensive review and 2016 update’
    I De Kouchkovsky, M Abdul-Hay
    Blood Cancer Journal.2016; 6(7): e441.     CrossRef
  • Recent updates of precision therapy for gastric cancer: Towards optimal tailored management
    Moon Kyung Joo, Jong-Jae Park, Hoon Jai Chun
    World Journal of Gastroenterology.2016; 22(19): 4638.     CrossRef
  • Targeted inhibition of STATs and IRFs as a potential treatment strategy in cardiovascular disease
    Malgorzata Szelag, Anna Piaszyk-Borychowska, Martyna Plens-Galaska, Joanna Wesoly, Hans A.R. Bluyssen
    Oncotarget.2016; 7(30): 48788.     CrossRef
  • Cell-cell and cell-matrix adhesion in survival and metastasis: Stat3 versus Akt
    Maximilian Niit, Victoria Hoskin, Esther Carefoot, Mulu Geletu, Rozanne Arulanandam, Bruce Elliott, Leda Raptis
    Biomolecular Concepts.2015; 6(5-6): 383.     CrossRef
  • Phase 1 and pharmacological trial of OPB‐31121, a signal transducer and activator of transcription‐3 inhibitor, in patients with advanced hepatocellular carcinoma
    Takuji Okusaka, Hideki Ueno, Masafumi Ikeda, Shuichi Mitsunaga, Masato Ozaka, Hiroshi Ishii, Osamu Yokosuka, Yoshihiko Ooka, Ryo Yoshimoto, Yasuo Yanagihara, Kiwamu Okita
    Hepatology Research.2015; 45(13): 1283.     CrossRef
  • STAT3 Inhibition Enhances the Therapeutic Efficacy of Immunogenic Chemotherapy by Stimulating Type 1 Interferon Production by Cancer Cells
    Heng Yang, Takahiro Yamazaki, Federico Pietrocola, Heng Zhou, Laurence Zitvogel, Yuting Ma, Guido Kroemer
    Cancer Research.2015; 75(18): 3812.     CrossRef
  • 18,343 View
  • 252 Download
  • 93 Web of Science
  • 93 Crossref
Close layer
Phase I Study of CKD-516, a Novel Vascular Disrupting Agent, in Patients with Advanced Solid Tumors
Do-Youn Oh, Tae-Min Kim, Sae-Won Han, Dong-Yeop Shin, Yun Gyoo Lee, Keun-Wook Lee, Jee Hyun Kim, Tae-You Kim, In-Jin Jang, Jong-Seok Lee, Yung-Jue Bang
Cancer Res Treat. 2016;48(1):28-36.   Published online February 23, 2015
DOI: https://doi.org/10.4143/crt.2014.258
AbstractAbstract PDFPubReaderePub
Purpose
CKD-516 is a newly developed vascular disrupting agent. This phase I dose-escalation study of CKD-516 was conducted to determine maximum-tolerated dose (MTD), safety, pharmacokinetics, and preliminary antitumor efficacy in patients with advanced solid tumors. Materials and Methods Patients received CKD-516 intravenously on D1 and D8 every 3 weeks, in a standard 3+3 design. Safety was evaluated by National Cancer Institute Common Terminology Criteria for Adverse Events ver. 4.02 and response was assessed by Response Evaluation Criteria in Solid Tumor ver. 1.1.
Results
Twenty-three patients were treated with CKD-516 at seven dosing levels: 1 mg/m2/day (n=3), 2 mg/m2/day (n=3), 3.3 mg/m2/day (n=3), 5 mg/m2/day (n=3), 7 mg/m2/day (n=3), 9 mg/m2/day (n=6), and 12 mg/m2/day (n=2). Mean age was 54 and 56.5% of patients were male. Two dose-limiting toxicities, which were both grade 3 hypertension, were observed in two patients at 12 mg/m2/day. The MTD was determined as 12 mg/m2/day. Most common adverse events were gastrointestinal adverse events (diarrhea, 34.8% [30.4% grade 1/2, 13.0% grade 3]; nausea, 21.7% [all grade 1/2]; vomiting, 21.7% [all grade 1/2]), myalgia (17.4%, all grade 1/2), and abdominal pain (21.7% [21.7% grade 1/2, 4.3% grade 3]). The pharmacokinetic study showed the dose-linearity of all dosing levels. Among 23 patients, six patients (26.1%) showed stable disease. Median progression-free survival was 39 days (95% confidence interval, 37 to 41 days). Conclusion This study demonstrates feasibility of CKD-516, novel vascular disrupting agent, in patients with advanced solid tumor. MTD of CKD-516 was defined as 12 mg/m2/day on D1 and D8 every 3 weeks.

Citations

Citations to this article as recorded by  
  • Natural‐Product‐Inspired Discovery of Trimethoxyphenyl‐1,2,4‐triazolosulfonamides as Potent Tubulin Polymerization Inhibitors
    Vajja Krishna Rao, Anvesh Ashtam, Dulal Panda, Sankar K. Guchhait
    ChemMedChem.2024;[Epub]     CrossRef
  • Thiazole, Isatin and Phthalimide Derivatives Tested in vivo against Cancer Models: A Literature Review of the Last Six Years
    Aline Ferreira Pinto, Janine Siqueira Nunes, José Eduardo Severino Martins, Amanda Calazans Leal, Carla Cauanny Vieira Costa Silva, Anderson José Firmino Santos da Silva, Daiane Santiago da Cruz Olímpio, Elineide Tayse Noberto da Silva, Thiers Araújo Camp
    Current Medicinal Chemistry.2024; 31(20): 2991.     CrossRef
  • CKD-516 potentiates the anti-cancer activity of docetaxel against epidermal growth factor receptor tyrosine kinase inhibitor-resistant lung cancer
    Soo Jin Kim, Kyunghyeon Lee, Jaewoo Park, Miso Park, U. Ji Kim, Se-mi Kim, Keun Ho Ryu, Keon Wook Kang
    Toxicological Research.2023; 39(1): 61.     CrossRef
  • Virulence-attenuated Salmonella engineered to secrete immunomodulators reduce tumour growth and increase survival in an autochthonous mouse model of breast cancer
    Lance B. Augustin, Liming Milbauer, Sara E. Hastings, Arnold S. Leonard, Daniel A. Saltzman, Janet L. Schottel
    Journal of Drug Targeting.2021; 29(4): 430.     CrossRef
  • A phase 1 dose-escalation and dose-expansion study to assess the safety and efficacy of CKD-516, a novel vascular disrupting agent, in combination with Irinotecan in patients with previously treated metastatic colorectal cancer
    Hyehyun Jeong, Yong Sang Hong, Jeong Eun Kim, Hyeong-Seok Lim, Joong Bae Ahn, Sang Joon Shin, Young Suk Park, Seung Tae Kim, Sae-Won Han, Tae-You Kim, Tae Won Kim
    Investigational New Drugs.2021; 39(5): 1335.     CrossRef
  • Application of triazoles as bioisosteres and linkers in the development of microtubule targeting agents
    M. Shaheer Malik, Saleh A. Ahmed, Ismail I. Althagafi, Mohammed Azam Ansari, Ahmed Kamal
    RSC Medicinal Chemistry.2020; 11(3): 327.     CrossRef
  • Tumor regression and potentiation of polymeric vascular disrupting therapy through reprogramming of a hypoxia microenvironment with temsirolimus
    Haiyang Yu, Na Shen, Yanli Bao, Li Chen, Zhaohui Tang
    Biomaterials Science.2020; 8(1): 325.     CrossRef
  • Phase I and pharmacokinetic study of the vascular‐disrupting agent CKD‐516 (NOV120401) in patients with refractory solid tumors
    Hark Kyun Kim, Jeong Won Kang, Young‐Whan Park, Jung Young Kim, Minchae Kim, Soo Jin Kim, Se‐mi Kim, Keun Ho Ryu, Seonghae Yoon, Yun Kim, Joo‐Youn Cho, Keun Seok Lee, Tak Yun, Kiwon Kim, Mi Hyang Kwak, Tae‐Sung Kim, Jinsoo Chung, Joong‐Won Park
    Pharmacology Research & Perspectives.2020;[Epub]     CrossRef
  • Revolutionizing the landscape of colorectal cancer treatment: The potential role of immune checkpoint inhibitors
    Mai F. Tolba
    International Journal of Cancer.2020; 147(11): 2996.     CrossRef
  • Discovery of tertiary amide derivatives incorporating benzothiazole moiety as anti-gastric cancer agents in vitro via inhibiting tubulin polymerization and activating the Hippo signaling pathway
    Jian Song, Qiu-Lei Gao, Bo-Wen Wu, Ting Zhu, Xin-Xin Cui, Cheng-Jun Jin, Shu-Yu Wang, Sheng-Hui Wang, Dong-Jun Fu, Hong-Min Liu, Sai-Yang Zhang, Yan-Bing Zhang, Yong-Chun Li
    European Journal of Medicinal Chemistry.2020; 203: 112618.     CrossRef
  • Anti-tumor efficacy of CKD-516 in combination with radiation in xenograft mouse model of lung squamous cell carcinoma
    Min-Young Kim, Jung-Young Shin, Jeong-Oh Kim, Kyoung-Hwa Son, Yeon Sil Kim, Chan Kwon Jung, Jin-Hyoung Kang
    BMC Cancer.2020;[Epub]     CrossRef
  • Discovery and optimization of 3,4,5-trimethoxyphenyl substituted triazolylthioacetamides as potent tubulin polymerization inhibitors
    Fang Yang, Cai-Ping He, Peng-Cheng Diao, Kwon Ho Hong, Jin-Jun Rao, Pei-Liang Zhao
    Bioorganic & Medicinal Chemistry Letters.2019; 29(1): 22.     CrossRef
  • Vascular Disrupting Agents in cancer treatment: Cardiovascular toxicity and implications for co-administration with other cancer chemotherapeutics
    Jason H. Gill, Kimberly L. Rockley, Carol De Santis, Asma K. Mohamed
    Pharmacology & Therapeutics.2019; 202: 18.     CrossRef
  • Combretastatin A4 Nanoparticles Combined with Hypoxia-Sensitive Imiquimod: A New Paradigm for the Modulation of Host Immunological Responses during Cancer Treatment
    Na Shen, Jing Wu, Chenguang Yang, Haiyang Yu, Shengcai Yang, Tete Li, Jingtao Chen, Zhaohui Tang, Xuesi Chen
    Nano Letters.2019; 19(11): 8021.     CrossRef
  • Recent advances in trimethoxyphenyl (TMP) based tubulin inhibitors targeting the colchicine binding site
    Ling Li, Sibo Jiang, Xiaoxun Li, Yao Liu, Jing Su, Jianjun Chen
    European Journal of Medicinal Chemistry.2018; 151: 482.     CrossRef
  • Combination of anti-vascular agent - DMXAA and HIF-1α inhibitor - digoxin inhibits the growth of melanoma tumors
    Ryszard Smolarczyk, Tomasz Cichoń, Ewelina Pilny, Magdalena Jarosz-Biej, Aleksandra Poczkaj, Natalia Kułach, Stanisław Szala
    Scientific Reports.2018;[Epub]     CrossRef
  • Chemoembolization with Vascular Disrupting Agent CKD-516 Dissolved in Ethiodized Oil in Combination with Doxorubicin: A VX2 Tumor Model Study
    In Joon Lee, Myungsu Lee, Soo Jin Kim, You Kyung Kim, Jong Yun Won, Jin Wook Chung
    Journal of Vascular and Interventional Radiology.2018; 29(8): 1078.     CrossRef
  • Enhanced efficacy of radiofrequency ablation for hepatocellular carcinoma using a novel vascular disrupting agent, CKD-516
    Su Jung Ham, YoonSeok Choi, Seul-I Lee, Jinil Kim, Young Il Kim, Jin Wook Chung, Kyung Won Kim
    Hepatology International.2017; 11(5): 446.     CrossRef
  • Early investigational tubulin inhibitors as novel cancer therapeutics
    Kunal Nepali, Ritu Ojha, Hsueh-Yun Lee, Jing-Ping Liou
    Expert Opinion on Investigational Drugs.2016; 25(8): 917.     CrossRef
  • Blocking Blood Flow to Solid Tumors by Destabilizing Tubulin: An Approach to Targeting Tumor Growth
    María-Jesús Pérez-Pérez, Eva-María Priego, Oskía Bueno, Maria Solange Martins, María-Dolores Canela, Sandra Liekens
    Journal of Medicinal Chemistry.2016; 59(19): 8685.     CrossRef
  • Current Advances of Tubulin Inhibitors in Nanoparticle Drug Delivery and Vascular Disruption/Angiogenesis
    Souvik Banerjee, Dong-Jin Hwang, Wei Li, Duane Miller
    Molecules.2016; 21(11): 1468.     CrossRef
  • 11,651 View
  • 139 Download
  • 21 Web of Science
  • 21 Crossref
Close layer
Phase I Study of Axitinib in Combination with Cisplatin and Capecitabine in Patients with Previously Untreated Advanced Gastric Cancer
Do-Youn Oh, Toshihiko Doi, Kuniaki Shirao, Keun-Wook Lee, Sook Ryun Park, Ying Chen, Liqiang Yang, Olga Valota, Yung-Jue Bang
Cancer Res Treat. 2015;47(4):687-696.   Published online February 12, 2015
DOI: https://doi.org/10.4143/crt.2014.225
AbstractAbstract PDFPubReaderePub
Purpose
This phase I trial evaluated the question of whether the standard starting dose of axitinib could be administered in combination with therapeutic doses of cisplatin/capecitabine in patients with previously untreated advanced gastric cancer, and assessed overall safety, pharmacokinetics, and preliminary antitumor activity of this combination.
Materials and Methods
Patients in dose level (DL) 1 received axitinib 5 mg twice a day (days 1 to 21) with cisplatin 80 mg/m2 (day 1) and capecitabine 1,000 mg/m2 twice a day (days 1 to 14) in 21-day cycles. Maximum tolerated dose (MTD) was the highest dose at which ≤ 30% of the first 12 patients experienced a dose-limiting toxicity (DLT) during cycle 1. Ten additional patients were enrolled and treated at the MTD in order to obtain additional safety and pharmacokinetic data.
Results
Three DLTs occurred during cycle 1 in three (25%) of the first 12 patients: ruptured abdominal aortic aneurysm, acute renal failure, and > 5 consecutive days of missed axitinib due to thrombocytopenia. DL1 was established as the MTD, since higher DL cohorts were not planned. Common grade 3/4 non-hematologic adverse events in 22 patients treated at DL1 included hypertension (36.4%) and decreased appetite and stomatitis (18.2% each). Cisplatin/capecitabine slightly increased axitinib exposure; axitinib decreased capecitabine and 5-fluorouracil exposure. Eight patients (36.4%) each had partial response or stable disease. Median response duration was 9.1 months; median progression-free survival was 3.8 months.
Conclusion
In patients with advanced gastric cancer, standard doses of axitinib plus therapeutic doses of cisplatin and capecitabine could be administered in combination. Adverse events were manageable.

Citations

Citations to this article as recorded by  
  • Construction of a hypoxia-immune-related prognostic panel based on integrated single-cell and bulk RNA sequencing analyses in gastric cancer
    Cuncan Deng, Guofei Deng, Hongwu Chu, Songyao Chen, Xiancong Chen, Xing Li, Yulong He, Chunhui Sun, Changhua Zhang
    Frontiers in Immunology.2023;[Epub]     CrossRef
  • Early TP53 Alterations Shape Gastric and Esophageal Cancer Development
    Pranshu Sahgal, Brandon M. Huffman, Deepa T. Patil, Walid K. Chatila, Rona Yaeger, James M. Cleary, Nilay S. Sethi
    Cancers.2021; 13(23): 5915.     CrossRef
  • High ELK3 Expression is Associated with the VEGF-C/VEGFR-3 Axis and Gastric Tumorigenesis and Enhances Infiltration of M2 Macrophages
    Wang Dazhi, Jiao Zheng, Ren Chunling
    Future Medicinal Chemistry.2020; 12(24): 2209.     CrossRef
  • RETRACTED: Long non-coding RNA LINC00978 promotes cell proliferation and tumorigenesis via regulating microRNA-497/NTRK3 axis in gastric cancer
    Ju-Yuan Bu, Wei-Ze Lv, Yi-Feng Liao, Xiao-Yu Xiao, Bao-Jun Lv
    International Journal of Biological Macromolecules.2019; 123: 1106.     CrossRef
  • RETRACTED ARTICLE: Anti-gastric cancer effect of Salidroside through elevating miR-99a expression
    Lin Yang, Yanan Yu, Qi Zhang, Xiaoyu Li, Cuiping Zhang, Tao Mao, Siliang Liu, Zibin Tian
    Artificial Cells, Nanomedicine, and Biotechnology.2019; 47(1): 3500.     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
  • Anti-angiogenic Therapy in Patients with Advanced Gastric and Gastroesophageal Junction Cancer: A Systematic Review
    Li-Tzong Chen, Do-Youn Oh, Min-Hee Ryu, Kun-Huei Yeh, Winnie Yeo, Roberto Carlesi, Rebecca Cheng, Jongseok Kim, Mauro Orlando, Yoon-Koo Kang
    Cancer Research and Treatment.2017; 49(4): 851.     CrossRef
  • Angiogenesis inhibitors in early development for gastric cancer
    Mauricio P. Pinto, Gareth I. Owen, Ignacio Retamal, Marcelo Garrido
    Expert Opinion on Investigational Drugs.2017; 26(9): 1007.     CrossRef
  • HER2-induced metastasis is mediated by AKT/JNK/EMT signaling pathway in gastric cancer
    Yiseul Choi, Young San Ko, Jin Ju Park, Youngsun Choi, Younghoon Kim, Jung-Soo Pyo, Bo Gun Jang, Douk Ho Hwang, Woo Ho Kim, Byung Lan Lee
    World Journal of Gastroenterology.2016; 22(41): 9141.     CrossRef
  • 15,075 View
  • 108 Download
  • 11 Web of Science
  • 9 Crossref
Close layer
Randomized Phase III Trial of Cisplatin, Epirubicin, Leucovorin, 5-Fluorouracil (PELF) Combination versus 5-fluorouracil Alone as Adjuvant Chemotherapy in Curative Resected Stage III Gastric Cancer
Jae Jin Lee, Si-Young Kim, Im sik Shin, Kyung Sam Cho, Hoong-Zae Joo, Choong Yoon, Yoon Wha Kim, Hwi Joong Yoon
Cancer Res Treat. 2004;36(2):140-145.   Published online April 30, 2004
DOI: https://doi.org/10.4143/crt.2004.36.2.140
AbstractAbstract PDFPubReaderePub
Purpose

The combination of cisplatin, epirubicin, leucovorin and 5-fluorouracil (PELF) administration, as adjuvant chemotherapy after curative resection for gastirc cancer, was compared with 5-fluorouracil (5-FU) administration alone. This paper reports the results of a prospective randomized comparison of the two regimens, PELF and 5-FU.

Methods

From August 1996 to July 1999, 54 patients were selected subsequent to being diagnosed with stage III cancer after a curative resection for gastric cancer. The patients were stratified according to stage IIIA/IIIB and subtotal/total gastrectomy, and then they were randomized into each treatment group, i.e. the PELF or 5-FU alone groups.

Results

54 assessable patients were enrolled in this study: 28 received PELF and 26 received 5-FU alone. 12 patients relapsed in each group and the median follow-up duration was 42 months (range: 10~77 months). The overall survival rate and disease-free survival rate (DFS) were not significantly different between two groups, (5-year survival of PELF vs. 5-FU: 57% vs. 64%, 5-year DFS: 54% vs. 51%). The PELF combination was more toxic in terms of anemia, anorexia, nausea and diarrhea than the 5-FU.

Conclusions

This study showed that the PELF combination, as an adjuvant therapy for gastric cancer after a curative resection, was a less effective treatment, and it had more toxic effects than the 5-FU.

Citations

Citations to this article as recorded by  
  • Multidisciplinary treatment strategy for locally advanced gastric cancer: A systematic review
    Kotaro Sugawara, Yoshikuni Kawaguchi, Yasuyuki Seto, Jean-Nicolas Vauthey
    Surgical Oncology.2021; 38: 101599.     CrossRef
  • The Efficacy and Safety of (Neo)Adjuvant Therapy for Gastric Cancer: A Network Meta-analysis
    Tom van den Ende, Emil ter Veer, Mélanie Machiels, Rosa M. A. Mali, Frank A. Abe Nijenhuis, Laura de Waal, Marety Laarman, Suzanne S. Gisbertz, Maarten C. C. M. Hulshof, Martijn G. H. van Oijen, Hanneke W. M. van Laarhoven
    Cancers.2019; 11(1): 80.     CrossRef
  • Prognostic and Predictive Factors for the Curative Treatment of Esophageal and Gastric Cancer in Randomized Controlled Trials: A Systematic Review and Meta-Analysis
    Tom van den Ende, Emil ter Veer, Rosa M. A. Mali, Mark I. van Berge Henegouwen, Maarten C. C. M. Hulshof, Martijn G. H. van Oijen, Hanneke W. M. van Laarhoven
    Cancers.2019; 11(4): 530.     CrossRef
  • COMplot, A Graphical Presentation of Complication Profiles and Adverse Effects for the Curative Treatment of Gastric Cancer: A Systematic Review and Meta-Analysis
    Tom van den Ende, Frank A. Abe Nijenhuis, Héctor G. van den Boorn, Emil ter Veer, Maarten C. C. M. Hulshof, Suzanne S. Gisbertz, Martijn G. H. van Oijen, Hanneke W. M. van Laarhoven
    Frontiers in Oncology.2019;[Epub]     CrossRef
  • Comparative effectiveness of adjuvant treatments for resected gastric cancer: a network meta-analysis
    Zhaolun Cai, Yiqiong Yin, Yuan Yin, Chaoyong Shen, Jian Wang, Xiaonan Yin, Zhixin Chen, Ye Zhou, Bo Zhang
    Gastric Cancer.2018; 21(6): 1031.     CrossRef
  • Current challenges of metastatic breast cancer
    Bora Lim, Gabriel N. Hortobagyi
    Cancer and Metastasis Reviews.2016; 35(4): 495.     CrossRef
  • Management of Gastroesophageal Junction Tumors
    Matthew P. Fox, Victor van Berkel
    Surgical Clinics of North America.2012; 92(5): 1199.     CrossRef
  • 9,589 View
  • 61 Download
  • 7 Crossref
Close layer
A Phase II Study of Genexol(R) (paclitaxel) in Metastatic Breast Cancer
Joo Young Jung, Hyun Chul Jeong, Sung Soo Yoon, Jae Hoon Lee, Jun Seok Kim, Hyo Jin Kim, Ki Hyun Kim, Jun O Park, Won Seop Lee, Dae Seog Heo, Yung Jue Bang, Noe Kyeong Kim
Cancer Res Treat. 2001;33(6):451-457.   Published online December 31, 2001
DOI: https://doi.org/10.4143/crt.2001.33.6.451
AbstractAbstract PDF
PURPOSE
Paclitaxel is a very effective agent in the treatment of breast cancer. Samyang Corporation has developed its own process to produce paclitaxel in a large volume using plant cell culture technology. To evaluate the efficacy and safety of Genexol(R) in patients with metastatic breast cancer who have failed to respond to standard therapy, we performed a prospective, multi- center phase II clinical trial.
MATERIALS AND METHODS
Patients with metastatic breast cancer were included in this study. Enrollees were required to have histologically confirmed breast cancer with bidimensionally measurable metastatic disease. Genexol(R) was administered at 175 mg/m2 as a 3-hour intravenous infusion every 3 weeks. All patients were premedicated with hydrocortisone, pheniramine maleate, and H2 blocker 30 minutes prior to paclitaxel. We planned to administer at least 4 courses of paclitaxel unless there was disease progression or unacceptable toxicity and to continue treatment up to a total of 6 courses in cases of objective response following 4 courses.
RESULTS
The median duration of follow-up was 8.9 (2.07~13.7) months. Forty-five patients were registered and 43 were eligible. The performance status of patients was ECOG 0~1 in 39 patients (90.7%) and 2 in 4 (9.3%). The location of metastases at the start of the study were the lung (15 patients), liver (8 patients), lymph nodes (22 patients), and other (7 patients). Among the 40 evaluable patients, 15 patients obtained partial responses (PRs) (37.5%, 95% CI: 22.5~52.5%). The median duration of response was 11.67 (4.1~11.7) months and the median time to progression was 7.73 (2.8~11.7) months. The median survival time was not reached at 13.7 months, and the overall survival rate at 13.7 months was 70.1%. The hematologic toxicity was primarily neutropenia with grade 3 or 4 in 10 patients (23.3%). The grade 3 or 4 non-hematologic toxicities included alopecia (17, 39.5%), myalgia (2, 4.7%), neuropathy (2, 4.7%), and pruritus (1, 2.3%). Mild hypersensitivity reaction was observed in 2 patients, although it did not cause withdrawal of the test drug.
CONCLUSION
The results suggest that the Genexol injection is an effective anticancer formulation for the treatment of metastatic breast cancer and toxicity is acceptable.

Citations

Citations to this article as recorded by  
  • Current status of nanomedicine in the chemotherapy of breast cancer
    A. I. Fraguas-Sánchez, C. Martín-Sabroso, A. Fernández-Carballido, A. I. Torres-Suárez
    Cancer Chemotherapy and Pharmacology.2019; 84(4): 689.     CrossRef
  • Association of the ABCB1 gene polymorphisms 2677G>T/A and 3435C>T with clinical outcomes of paclitaxel monotherapy in metastatic breast cancer patients
    H. Chang, S.Y. Rha, H.-C. Jeung, C.-K. Im, J.B. Ahn, W.S. Kwon, N.C. Yoo, J.K. Roh, H.C. Chung
    Annals of Oncology.2009; 20(2): 272.     CrossRef
  • 5,067 View
  • 41 Download
  • 2 Crossref
Close layer
Phase I Clinical Trial of Paclitaxel Plus Ifosfamide for the Patients with Refractory Ovarian Cancer
Ho Sawk Saw, Jae Kwan Lee
J Korean Cancer Assoc. 2000;32(5):895-903.
AbstractAbstract PDF
PURPOSE
Patients with advanced ovarian carcinoma and refractory to platinum based chemotherapy have a very poor prognosis and effective salvage regimens are needed. This study was conducted in order to determine the maximum tolerated dose (MTD) and dose limiting toxicity of combination with paclitaxel and ifosfamide.
MATERIALS AND METHODS
After premedication, patients received paclitaxel (110~225 mg/m2) as a 24 hour IV infusion on day 1. Ifosfamide (1,000~1,500 mg/m2) was given as a 12 hour IV infusion with standard dose of mesna on day 2~6. All patients received G-CSF (granulocyte colony stimulating factor) on day 6~15.
RESULTS
12 patients with advanced ovarian cancer entered this trial. Toxicity included bone marrow suppression, neuromuscular toxicity, urothelial toxicity, gastrointestinal toxicity, which occurred in 84.6%, 65.3%, 30.7%, 88.4% of cycles.
CONCLUSION
Neuromuscular toxicity was dose limiting toxicity. Maximum tolerated dose in com bination with paclitaxel and ifosfamide was 175 mg/m2 of paclitaxel and 1,500 mg/m2 of ifosfamide.
  • 2,077 View
  • 17 Download
Close layer
Phase II Multicenter Trial of Paclitaxel for Metastatic Breast Cancer
Jae Ho Byun, Sung Soo Yoon, Chan Hyung Park, Sung Rok Kim, Si Young Kim, Hyo Jin Kim, Soo Mee Bang, Heung Moon Chang, Dae Seog Heo, Yung Jue Bang, Noe Kyeong Kim
J Korean Cancer Assoc. 2000;32(3):545-552.
AbstractAbstract PDF
PURPOSE
To evaluate the efficacy and safety of paclitaxel for metastatic breast cancer patients who had received previous chemotherapy, we have performed phase II multicenter trial between December 1997 and December 1998.
MATERIALS AND METHOD
Thirty patients were accrued to this study and paclitaxel was administered at 175 mg/m2 as a 3-hour intravenous infusion every 3 weeks until the progression of the disease.
RESULTS
Objective response were observed in 13 of 30 patients (43.3%). There were 1 complete response (3.3%) and 12 partial responses (40%). Especially, 50% (11/22) of patients who had received prior anthracycline-containing regimens for adjuvant or metastatic disease responded to paclitaxel. Responses were observed in all sites of metastatic disease. One hundred forty-nine cycles of treatment were administered, with a median of six cycles per patient. Grade III and IV toxicities included neutropenia (24%), elevated liver enzyme (10%), peripheral neuropathy (10%), arthralgia/myalgia (23%), and alopecia (87%). No significant hypersensitivity type reaction or cardiac arrhythmia were seen. Median duration of response was 7.2 months.
CONCLUSIONS
These results suggested that paclitaxel is active therapeutic agent in metastatic breast cancer patients and it can be safely administered by 3-hour intravenous infusion with premedication.
  • 2,902 View
  • 37 Download
Close layer
Clinical Trial
Efficacy of Gemcitabine Chemotherpy in Advanced Non-small cell Lung Cancer ( NSCLC ): A Phase 2 Study
Hyuk Jae Chang, Joong Bae Ahn, Jun Gu Lee, Kwang Yong Shim, Sun Young Rha, Sae Kyu Kim, Jun Chang, Sung Kyu Kim, Won Young Lee, Nae Chun Yoo, Hyun Cheol Chung, Jae Kyung Roh, Byung Soo Kim, Sung Jun Choi, Tae Won Kim, Chul Won Suh, Joo Hang Kim
J Korean Cancer Assoc. 1999;31(3):523-532.
AbstractAbstract PDF
PURPOSE
To evaluate the efficacy and safety of gemcitabine, a pyrimidine antimetabolite against advanced non-small cell lung cancer (NSCLC).
MATERIALS AND METHODS
Forty patients with unresectable stage IIIb to IV, pathologacally documented NSCLC were evaluated. Patients received gemcitabine 1000 mg/m, as a 30 to 60-min, intravenous infusion on days 1, 8 and 15, which was repeated every 28 days. Responses were assessed every two courses. Twenty-five to fifty percent dose reduction was permitted, ptovided that overall toxicity was severe according to World Health Organization (WHO) toxicity criteria.
RESULTS
Of all 40 patients (32 men, 8 women; age range 37 to 73 years; median 63 years), 3S patients were assessable for response. 15 patients had stage IIIb disease and 25 had stage IV. Nineteen patients were histologically classified as adenocarcinoma (47.5%), 17 as squamous cell carcinoma (42.5%), 1 as large cell carcinoma (2.5%), 1 as mixed carcinoma (2.5%) and 2 as undifferentiated carcinoma (5.0%). The overall response rate was 20%. None of the patients showed complete response while 7 showed partial response (20%), 5 had stable diseases (23%) and 23 had progressive diseases (57%). During a total of 119 courses, hematologic toxicity was negligible. Granulo- cytopenia worse than WHO grade 3 occured in 11.8%, anemia in O.S% and thrombocytopenia in 0.8%, respectively. Non-hematologic toxicity was minor and easily controlled. There was no case of febrile neutropenia or treatment-related death.
CONCLUSION
The single agent efficacy of gemcitabine is comparable to other agents commonly used to treat NSCLC. Gemcitabine has unusually mild side effect profile for such an active agent. This significant activity in conjunction with a very favorable toxicity profile supports further investigation in combination with other agents in patients with inoperable NSCLC.
  • 3,876 View
  • 35 Download
Close layer

Cancer Res Treat : Cancer Research and Treatment
Close layer
TOP