1
|
Zheng K, Jin G, Cao R, Gao Y, Xu J, Chai R, Kang Y. Targeting on the PI3K/mTOR: a potential treatment strategy for clear cell ovarian carcinoma. Cancer Chemother Pharmacol 2025; 95:21. [PMID: 39792198 PMCID: PMC11723846 DOI: 10.1007/s00280-024-04748-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/29/2024] [Indexed: 01/12/2025]
Abstract
PURPOSE Ovarian clear cell carcinoma is a highly malignant gynecological tumor characterized by a high rate of chemotherapy resistance and poor prognosis. The PI3K/AKT/mTOR pathway is well-known to be closely related to the progression of various malignancies, and recent studies have indicated that this pathway may play a critical role in the progression and worsening of OCCC. METHODS In this study, we investigated the combined effects of WX390, a dual inhibitor of PI3K/mTOR, and cisplatin on OCCC through both in vitro and in vivo experiments to further elucidate their therapeutic effects. RESULTS WX390 significantly inhibited the proliferation of human OCCC cell lines ES2 and OVISE, while promoting apoptosis. Furthermore, the combination of WX390 with CDDP exhibited a synergistic effect, markedly increasing the sensitivity of OCCC cells to chemotherapeutic agents and significantly suppressing tumor growth in PDX models. Western blot and RNA-seq analyses revealed that WX390 robustly inhibited the PI3K/AKT/mTOR pathway, interrupt autophagy, altered cell cycle dynamics, and induced apoptosis. CONCLUSION This study comprehensively assessed the efficacy of WX390 across multiple models of OCCC, laying a solid foundation for the development of new therapeutic strategies for this challenging malignancy.
Collapse
Affiliation(s)
- Kewei Zheng
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Guanqin Jin
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Rui Cao
- Department of Gynecology, Dalian Obstetrics and Gynecology Hospital, Dalian, 116033, China
| | - Yi Gao
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Jing Xu
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Ranran Chai
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Yu Kang
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China.
| |
Collapse
|
2
|
Liu Z, Jing C, Kong F. From clinical management to personalized medicine: novel therapeutic approaches for ovarian clear cell cancer. J Ovarian Res 2024; 17:39. [PMID: 38347608 PMCID: PMC10860311 DOI: 10.1186/s13048-024-01359-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 01/26/2024] [Indexed: 02/15/2024] Open
Abstract
Ovarian clear-cell cancer is a rare subtype of epithelial ovarian cancer with unique clinical and biological features. Despite optimal cytoreductive surgery and platinum-based chemotherapy being the standard of care, most patients experience drug resistance and a poor prognosis. Therefore, novel therapeutic approaches have been developed, including immune checkpoint blockade, angiogenesis-targeted therapy, ARID1A synthetic lethal interactions, targeting hepatocyte nuclear factor 1β, and ferroptosis. Refining predictive biomarkers can lead to more personalized medicine, identifying patients who would benefit from chemotherapy, targeted therapy, or immunotherapy. Collaboration between academic research groups is crucial for developing prognostic outcomes and conducting clinical trials to advance treatment for ovarian clear-cell cancer. Immediate progress is essential, and research efforts should prioritize the development of more effective therapeutic strategies to benefit all patients.
Collapse
Affiliation(s)
- Zesi Liu
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Dalian Medical University, Dalian, 116000, Liaoning Province, China
| | - Chunli Jing
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116000, Liaoning Province, China
| | - Fandou Kong
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Dalian Medical University, Dalian, 116000, Liaoning Province, China.
| |
Collapse
|
3
|
Kao TW, Bai GH, Wang TL, Shih IM, Chuang CM, Lo CL, Tsai MC, Chiu LY, Lin CC, Shen YA. Novel cancer treatment paradigm targeting hypoxia-induced factor in conjunction with current therapies to overcome resistance. J Exp Clin Cancer Res 2023; 42:171. [PMID: 37460927 DOI: 10.1186/s13046-023-02724-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/29/2023] [Indexed: 07/20/2023] Open
Abstract
Chemotherapy, radiotherapy, targeted therapy, and immunotherapy are established cancer treatment modalities that are widely used due to their demonstrated efficacy against tumors and favorable safety profiles or tolerability. Nevertheless, treatment resistance continues to be one of the most pressing unsolved conundrums in cancer treatment. Hypoxia-inducible factors (HIFs) are a family of transcription factors that regulate cellular responses to hypoxia by activating genes involved in various adaptations, including erythropoiesis, glucose metabolism, angiogenesis, cell proliferation, and apoptosis. Despite this critical function, overexpression of HIFs has been observed in numerous cancers, leading to resistance to therapy and disease progression. In recent years, much effort has been poured into developing innovative cancer treatments that target the HIF pathway. Combining HIF inhibitors with current cancer therapies to increase anti-tumor activity and diminish treatment resistance is one strategy for combating therapeutic resistance. This review focuses on how HIF inhibitors could be applied in conjunction with current cancer treatments, including those now being evaluated in clinical trials, to usher in a new era of cancer therapy.
Collapse
Affiliation(s)
- Ting-Wan Kao
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110301, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 110301, Taiwan
| | - Geng-Hao Bai
- Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei City, 100225, Taiwan
| | - Tian-Li Wang
- Departments of Pathology, Oncology and Gynecology and Obstetrics, Johns Hopkins Medical Institutions, 1550 Orleans StreetRoom 306, Baltimore, MD, CRB221231, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ie-Ming Shih
- Departments of Pathology, Oncology and Gynecology and Obstetrics, Johns Hopkins Medical Institutions, 1550 Orleans StreetRoom 306, Baltimore, MD, CRB221231, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chi-Mu Chuang
- Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei, 112304, Taiwan
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, 112201, Taiwan
- Department of Midwifery and Women Health Care, National Taipei University of Nursing and Health Sciences, Taipei, 112303, Taiwan
| | - Chun-Liang Lo
- Department of Biomedical Engineering, National Yang-Ming Chiao Tung University, Taipei, 112304, Taiwan
- Medical Device Innovation and Translation Center, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
| | - Meng-Chen Tsai
- Department of General Medicine, Taipei Medical University Hospital, Taipei, 110301, Taiwan
| | - Li-Yun Chiu
- Department of General Medicine, Mackay Memorial Hospital, Taipei, 104217, Taiwan
| | - Chu-Chien Lin
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110301, Taiwan
- School of Medicine, College of Medicine, Taipei Medical University, Taipei City, 110301, Taiwan
| | - Yao-An Shen
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110301, Taiwan.
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 110301, Taiwan.
- International Master/Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, 110301, Taiwan.
| |
Collapse
|
4
|
Muthamil S, Kim HY, Jang HJ, Lyu JH, Shin UC, Go Y, Park SH, Lee HG, Park JH. Understanding the relationship between cancer associated cachexia and hypoxia-inducible factor-1. Biomed Pharmacother 2023; 163:114802. [PMID: 37146421 DOI: 10.1016/j.biopha.2023.114802] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/07/2023] Open
Abstract
Cancer-associated cachexia (CAC) is a multifactorial disorder characterized by an unrestricted loss of body weight as a result of muscle and adipose tissue atrophy. Cachexia is influenced by several factors, including decreased metabolic activity and food intake, an imbalance between energy uptake and expenditure, excessive catabolism, and inflammation. Cachexia is highly associated with all types of cancers responsible for more than half of cancer-related mortalities worldwide. In healthy individuals, adipose tissue significantly regulates energy balance and glucose homeostasis. However, in metastatic cancer patients, CAC occurs mainly because of an imbalance between muscle protein synthesis and degradation which are organized by certain extracellular ligands and associated signaling pathways. Under hypoxic conditions, hypoxia-inducible factor-1 (HIF-1α) accumulated and translocated to the nucleus and activate numerous genes involved in cell survival, invasion, angiogenesis, metastasis, metabolic reprogramming, and cancer stemness. On the other hand, the ubiquitination proteasome pathway is inhibited during low O2 levels which promote muscle wasting in cancer patients. Therefore, understanding the mechanism of the HIF-1 pathway and its metabolic adaptation to biomolecules is important for developing a novel therapeutic method for cancer and cachexia therapy. Even though many HIF inhibitors are already in a clinical trial, their mechanism of action remains unknown. With this background, this review summarizes the basic concepts of cachexia, the role of inflammatory cytokines, pathways connected with cachexia with special reference to the HIF-1 pathway and its regulation, metabolic changes, and inhibitors of HIFs.
Collapse
Affiliation(s)
- Subramanian Muthamil
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Hyun Yong Kim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Hyun-Jun Jang
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Ji-Hyo Lyu
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Ung Cheol Shin
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Younghoon Go
- Korean Medicine (KM)-application Center, Korea Institute of Oriental Medicine, Daegu, Republic of Korea
| | - Seong-Hoon Park
- Genetic and Epigenetic Toxicology Research Group, Korea Institute of Toxicology, Daejeon 34141, Republic of Korea
| | - Hee Gu Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Jun Hong Park
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea; University of Science & Technology (UST), KIOM campus, Korean Convergence Medicine Major, Daejeon 34054, Republic of Korea.
| |
Collapse
|
5
|
Saini U, Smith BQ, Dorayappan KDP, Yoo JY, Maxwell GL, Kaur B, Konishi I, O’Malley D, Cohn DE, Selvendiran K. Targeting TMEM205 mediated drug resistance in ovarian clear cell carcinoma using oncolytic virus. J Ovarian Res 2022; 15:130. [PMID: 36476493 PMCID: PMC9730683 DOI: 10.1186/s13048-022-01054-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 10/17/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Ovarian clear cell carcinoma (OCCC) accounts for approximately 8-10% of epithelial ovarian cancers in the United States. Although it is rare, OCCC usually presents with treatment challenges and the overall prognosis is far worse than high grade serous ovarian cancer HGSOC. The objective of this study was to examine the therapeutic relevance of combining oncolytic virus with cisplatin for ovarian cancer clear cell carcinoma (OCCC). RESULTS We identified that TMEM205, a recently discovered transmembrane protein, contributes to chemoresistance in OCCC cells via the exosomal pathway. Mechanistically, TMEM205 undergoes ligand-independent constitutive endocytosis and co-localizes with Rab11 to contribute to the late recycling endosomes in a clathrin-independent manner. Further, we observed that oncolytic virus (oHSV) pretreatment followed by treatment with cisplatin decreases TMEM205 expression and sensitizes cells to cisplatin in a synergistic manner in OCCC cells. TMEM205 interacts with glycoprotein-C of oHSV post-infection; both of these proteins undergo ubiquitination and ultimately get shuttled outside the cell via exosomes. Thus, we demonstrate the mechanotransduction pathway of TMEM205-mediated chemoresistance along with targeting this pathway using oHSV and cisplatin as a powerful therapeutic strategy for OCCC. oHSV combination with cisplatin inhibits OCCC tumor growth in vivo in immunodeficient and immunocompetent mice models. CONCLUSION Our results suggest that the combination of oHSV and cisplatin in immunocompetent as well as immune deficient OCCC tumor bearing mice reduces overall tumor burden as well as metastatic disease thereby providing survival benefit. Additionally, the detection of TMEM205 in exosomal cargo early in OCCC development has potential to be exploited as a biomarker.
Collapse
Affiliation(s)
- Uksha Saini
- grid.412332.50000 0001 1545 0811Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - Brentley Q. Smith
- grid.412332.50000 0001 1545 0811Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - Kalpana Deepa Priya Dorayappan
- grid.412332.50000 0001 1545 0811Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - Ji Young Yoo
- grid.267308.80000 0000 9206 2401Department of Neurosurgery, University of Texas, Health Science Center, Houston, USA
| | - G. Larry Maxwell
- grid.414629.c0000 0004 0401 0871Inova Women’s Service Line and the Inova Schar Cancer Institute, Falls Church, VA USA
| | - Balveen Kaur
- grid.267308.80000 0000 9206 2401Department of Neurosurgery, University of Texas, Health Science Center, Houston, USA
| | - Ikuo Konishi
- grid.258799.80000 0004 0372 2033Division of GYN/ONC, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - David O’Malley
- grid.412332.50000 0001 1545 0811Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - David E. Cohn
- grid.412332.50000 0001 1545 0811Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - Karuppaiyah Selvendiran
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA.
| |
Collapse
|
6
|
Farley JH, Brady WE, O'Malley D, Fujiwara K, Yonemori K, Bonebrake A, Secord AA, Stephan JM, Walker JL, Nam JH, Birrer MJ, Gershenson DM. A phase II evaluation of temsirolimus with carboplatin and paclitaxel followed by temsirolimus consolidation in clear cell ovarian cancer: An NRG oncology trial. Gynecol Oncol 2022; 167:423-428. [PMID: 36244829 PMCID: PMC9789681 DOI: 10.1016/j.ygyno.2022.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVE The primary objective of the study was to estimate the 12-month progression-free survival (PFS) for carboplatin/paclitaxel + temsirolimus in women with newly diagnosed clear cell ovarian cancer (CCOC), compared to historical controls in this patient population. METHODS Patients with Stage III or IV CCOC were treated with Paclitaxel 175 mg/m2 on Day 1, Carboplatin AUC 6 Day 1, and temsirolimus (CCI-779) 25 mg IV Days 1 and 8 every 3 weeks for Cycles 1-6 or disease progression, followed by consolidation therapy with temsirolimus 25 mg Days 1, 8, and 15 every 3 weeks cycles 7-17 or until disease progression. RESULTS Ninety patients were accrued to the study: 45 in the US and Korea (US/Korea) and 45 in Japan. Twenty-two percent received ≤6 cycles of therapy while 28% completed all 17 cycles of chemotherapy. Median PFS (OS) was 11 (23) months for US/Korea and 12 (26) months for Japan. In the US, none of suboptimally debulked patients had PFS >12 months, and 49% of optimal patients did, compared to 25% and 59% in Japan. Most common grade 3-4 adverse events were neutropenia, leukopenia, anemia, thrombocytopenia, hypertension, hypertriglyceridemia, and oral mucositis. CONCLUSION The carboplatin/paclitaxel + temsirolimus regimen was well tolerated. In optimally debulked patients, 54% had a PFS >12 months. This regimen did not statistically significantly increase PFS at 12 months compared to historical controls. No statistically significant differences in PFS or OS were observed between US/Korea vs Japan, or Asians vs non-Asians.
Collapse
Affiliation(s)
- John H Farley
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Creighton University School of Medicine at St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA.
| | - William E Brady
- NRG Oncology, Clinical Trial Development Division, Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | | | | | - Kan Yonemori
- National Cancer Center Hospital, 5 Chome-1 - 1 Tsukiji, Chuo City, Tokyo 104-0045, Japan.
| | - Albert Bonebrake
- Cancer Research for the Ozarks-Cox Health, Springfield, MO, USA.
| | | | | | - Joan L Walker
- University of Oklahoma, Oklahoma City, OK 73104, USA.
| | - Joo-Hyun Nam
- Asan Medical Center, University of Ulsan, Seoul 13876 05505, KR, Republic of Korea.
| | | | - David M Gershenson
- Dept. of Gynecologic Oncology, University of Texas, MD, USA; Anderson Cancer Center, Unit 1362, PO Box 301439, Houston, TX 77230-1439, USA.
| |
Collapse
|
7
|
Zhao Y, Li J, Chen J, Ye M, Jin X. Functional roles of E3 ubiquitin ligases in prostate cancer. J Mol Med (Berl) 2022; 100:1125-1144. [PMID: 35816219 DOI: 10.1007/s00109-022-02229-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 12/16/2022]
Abstract
Prostate cancer (PCa) is a malignant epithelial tumor of the prostate gland with a high male cancer incidence. Numerous studies indicate that abnormal function of ubiquitin-proteasome system (UPS) is associated with the progression and metastasis of PCa. E3 ubiquitin ligases, key components of UPS, determine the specificity of substrates, and substantial advances of E3 ubiquitin ligases have been reached recently. Herein, we introduce the structures and functions of E3 ubiquitin ligases and summarize the mechanisms of E3 ubiquitin ligases-related PCa signaling pathways. In addition, some progresses in the development of inhibitors targeting E3 ubiquitin ligases are also included.
Collapse
Affiliation(s)
- Yiting Zhao
- Department of Oncology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, 315020, China.,Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, 315211, China.,Department of Chemoradiotherapy, the Affiliated People's Hospital of Ningbo University, Ningbo, 315040, China
| | - Jinyun Li
- Department of Oncology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, 315020, China.,Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, 315211, China
| | - Jun Chen
- Department of Chemoradiotherapy, the Affiliated People's Hospital of Ningbo University, Ningbo, 315040, China
| | - Meng Ye
- Department of Oncology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, 315020, China.,Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, 315211, China
| | - Xiaofeng Jin
- Department of Oncology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, 315020, China. .,Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
8
|
de Keijzer MJ, de Klerk DJ, de Haan LR, van Kooten RT, Franchi LP, Dias LM, Kleijn TG, van Doorn DJ, Heger M. Inhibition of the HIF-1 Survival Pathway as a Strategy to Augment Photodynamic Therapy Efficacy. Methods Mol Biol 2022; 2451:285-403. [PMID: 35505024 DOI: 10.1007/978-1-0716-2099-1_19] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Photodynamic therapy (PDT) is a non-to-minimally invasive treatment modality that utilizes photoactivatable drugs called photosensitizers to disrupt tumors with locally photoproduced reactive oxygen species (ROS). Photosensitizer activation by light results in hyperoxidative stress and subsequent tumor cell death, vascular shutdown and hypoxia, and an antitumor immune response. However, sublethally afflicted tumor cells initiate several survival mechanisms that account for decreased PDT efficacy. The hypoxia inducible factor 1 (HIF-1) pathway is one of the most effective cell survival pathways that contributes to cell recovery from PDT-induced damage. Several hundred target genes of the HIF-1 heterodimeric complex collectively mediate processes that are involved in tumor cell survival directly and indirectly (e.g., vascularization, glucose metabolism, proliferation, and metastasis). The broad spectrum of biological ramifications culminating from the activation of HIF-1 target genes reflects the importance of HIF-1 in the context of therapeutic recalcitrance. This chapter elaborates on the involvement of HIF-1 in cancer biology, the hypoxic response mechanisms, and the role of HIF-1 in PDT. An overview of inhibitors that either directly or indirectly impede HIF-1-mediated survival signaling is provided. The inhibitors may be used as pharmacological adjuvants in combination with PDT to augment therapeutic efficacy.
Collapse
Affiliation(s)
- Mark J de Keijzer
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Department of Pharmaceutics, Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Daniel J de Klerk
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Lianne R de Haan
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Robert T van Kooten
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Leonardo P Franchi
- Departamento de Bioquímica e Biologia Molecular, Instituto de Ciências Biológicas (ICB) 2, Universidade Federal de Goiás (UFG), Goiânia, GO, Brazil
- Faculty of Philosophy, Sciences, and Letters of Ribeirão Preto, epartment of Chemistry, Center of Nanotechnology and Tissue Engineering-Photobiology and Photomedicine Research Group,University of São Paulo, São Paulo, Brazil
| | - Lionel M Dias
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Tony G Kleijn
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Diederick J van Doorn
- Department of Gastroenterology and Hepatology, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Michal Heger
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China.
- Department of Pharmaceutics, Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
9
|
Ikeda H, Kakeya H. Targeting hypoxia-inducible factor 1 (HIF-1) signaling with natural products toward cancer chemotherapy. J Antibiot (Tokyo) 2021; 74:687-695. [PMID: 34331027 DOI: 10.1038/s41429-021-00451-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 05/29/2021] [Accepted: 06/04/2021] [Indexed: 02/06/2023]
Abstract
Tumor cells are often exposed to hypoxia because of the lower oxygen supply deep inside the tumor tissues. However, tumor cells survive in these severe conditions by adapting to hypoxic stress through the induction of hypoxia-inducible factor 1 (HIF-1) signaling. HIF-1 activation is responsible for the expression of numerous HIF-1 target genes, which are related to cell survival, proliferation, angiogenesis, invasion, metastasis, cancer stemness, and metabolic reprogramming. Therefore, HIF-1 is expected to be a potential pharmacological target for cancer therapy. Small molecules derived from natural products (microbial origin, plant-derived, or marine organisms) have been shown to have unique chemical structures and biological activities, including HIF-1 inhibition. Several studies identified HIF-1 inhibitors from natural products. In this review, we summarize the current HIF-1 signaling inhibitors originating from natural products with a variety of modes of action, mainly focusing on microbial metabolites.
Collapse
Affiliation(s)
- Hiroaki Ikeda
- Department of System Chemotherapy and Molecular Sciences, Division of Bioinformatics and Chemical Genomics, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Hideaki Kakeya
- Department of System Chemotherapy and Molecular Sciences, Division of Bioinformatics and Chemical Genomics, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, 606-8501, Japan.
| |
Collapse
|
10
|
Kobayashi H, Imanaka S. Toward an understanding of tissue factor pathway inhibitor-2 as a novel serodiagnostic marker for clear cell carcinoma of the ovary. J Obstet Gynaecol Res 2021; 47:2978-2989. [PMID: 34184357 DOI: 10.1111/jog.14916] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/16/2021] [Accepted: 06/20/2021] [Indexed: 12/19/2022]
Abstract
AIMS Tissue factor pathway inhibitor (TFPI)-2 has recently emerged as a serodiagnostic marker for patients with epithelial ovarian cancer (EOC), especially clear cell carcinoma (CCC). This review discusses the biological properties of TFPI-2 and why serum levels are elevated in CCC patients. METHODS A comprehensive literature search was conducted in PubMed up until March, 2021. RESULTS TFPI-2 is a Kunitz-type protease inhibitor and negatively regulates the enzymatic activities, such as plasmin. TFPI-2 has been characterized as a tumor suppressor gene and was frequently downregulated through promoter hypermethylation in various human cancers. In contrast, TFPI-2 was overexpressed only in CCC. TFPI-2 may be involved in the pathophysiology of CCC, possibly through regulation of coagulation system, stabilization of extracellular matrix (ECM), and induction of intracellular signal transduction. TFPI-2 suppresses tissue factor-induced hypercoagulation in a hypoxic environment. TFPI-2, secreted by CCC cells, platelets, and adjacent vascular endothelial cells, may suppress tumor growth and invasion through ECM remodeling. Nuclear TFPI-2 may suppress matrix metalloproteinase production via transcription factors and modulate caspase-mediated cell apoptosis. CCC cells may upregulate the TFPI-2 expression to adapt to survival in the demanding environment. TFPI-2 is secreted by CCC cells and enters the systemic circulation, resulting in elevated blood levels. DISCUSSION Serum TFPI-2 reflects the overexpression of TFPI-2 in CCC tissues and is a potential serodiagnostic marker. Further research is needed to explore the expression, clinical significance, biological function, and potential mechanism of TFPI-2 in CCC.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara, Japan.,Ms. Clinic MayOne, Kashihara, Nara, Japan
| | - Shogo Imanaka
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara, Japan.,Ms. Clinic MayOne, Kashihara, Nara, Japan
| |
Collapse
|
11
|
Xiao X, Liang S, Zhao Y, Huang D, Xing B, Cheng Z, Lin J. Core-shell structured 5-FU@ZIF-90@ZnO as a biodegradable nanoplatform for synergistic cancer therapy. NANOSCALE 2020; 12:3846-3854. [PMID: 31995084 DOI: 10.1039/c9nr09869k] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
High treatment efficiency and low drug toxicity are two key factors in tumor therapy. The development of multifunctional drug carrier systems is of great significance for the diagnosis and therapy of tumors. Herein, a novel biodegradable treatment system based on zeolitic imidazolate framework-90 (ZIF-90) was designed in this study. This 5-FU@ZIF-90@ZnO (FZZ) drug delivery system achieves synergistic treatment with antineoplastic 5-fluorouracil (5-FU) and zinc oxide, and also has good dispersibility in the acidic tumor microenvironment (TME), which enables the drug to achieve pH-controlled delivery in acidic organisms. Interestingly, zinc oxide nanoparticles can play a dual role here. They can prevent the premature leakage of drugs under physiological conditions. Moreover, Zn2+ produced after the decomposition of nanoparticles can act as a therapeutic agent, overcoming the tumor resistance to 5-FU and regulating a series of physiological reactions to inhibit tumor growth. It is worth noting that the porous ZIF-90 is an emerging drug carrier with a relatively high drug loading rate of 39% in this study. Synergistic 5-FU and ZnO nanoparticles have achieved tumor inhibition and have shown high therapeutic biosafety. Thus, the FZZ core-shell nanoparticles are a potential pH-controlled drug release system that can be applied to tumor treatment.
Collapse
Affiliation(s)
- Xiao Xiao
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China. and University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Shuang Liang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China. and University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Yajie Zhao
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China. and University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Dayu Huang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China.
| | - Bengang Xing
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University, Singapore 637371
| | - Ziyong Cheng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China. and University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China. and University of Science and Technology of China, Hefei, 230026, P. R. China
| |
Collapse
|
12
|
Precision medicine for ovarian clear cell carcinoma based on gene alterations. Int J Clin Oncol 2020; 25:419-424. [PMID: 32020380 DOI: 10.1007/s10147-020-01622-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 12/30/2019] [Indexed: 02/07/2023]
Abstract
Ovarian clear cell carcinoma (OCCC) is a histological subtype of epithelial ovarian carcinoma prevalent in Asians. No clear therapeutic selection based on molecular profile has been implemented for this disease. Oncogenic PIK3CA mutation, which activates the PIK3CA/AKT/mTOR signaling pathway, is a promising druggable alteration in OCCC. Recent studies by our group and others have identified the ARID1A mutation as another alteration linked to therapeutic selection based on synthetic lethality: deleterious ARID1A mutations, resulting in ARID1A deficiency, make OCCC cells sensitive to drugs targeting poly (ADP-ribose) polymerase and EZH2, as well as to glutathione inhibitors. In addition, we recently obtained evidence that ARID1A-deficient OCCC could benefit from gemcitabine treatment. Precision medicine based on gene alteration profiling might improve the prognosis of OCCC patients.
Collapse
|
13
|
Lu E, Li C, Wang J, Zhang C. Inflammation and angiogenesis in the corpus luteum. J Obstet Gynaecol Res 2019; 45:1967-1974. [PMID: 31373134 DOI: 10.1111/jog.14076] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/15/2019] [Indexed: 12/15/2022]
Abstract
Angiogenesis is a very important process that helps establish and maintain the normal structure and function of the corpus luteum (CL). Early luteal development can be considered a kind of physiological injury with an inflammatory response; therefore, the inflammatory response may play an important role in the luteal angiogenesis. The inflammatory response is companied by activated leukocytes and their mediators. For luteal tissue, numerous activated leukocytes such as macrophages, neutrophils and eosinophils are present in the early luteal phase and are widely involved in neovascularization. The objective of this review is to describe the role of the inflammatory factors in the angiogenesis and to discuss their mechanism. Knowledge of action and mechanism of these inflammatory factors on angiogenic activity will be beneficial for the understanding of luteal function.
Collapse
Affiliation(s)
- Enhang Lu
- Joint Programme of Nanchang University and Queen Mary University of London, School of Medicine, Nanchang University, Nanchang, China
| | - Chunjie Li
- Forth Clinical College, School of Medicine, Nanchang University, Nanchang, China
| | - Jing Wang
- Department of Microbiology, School of Medicine, Nanchang University, Nanchang, China
| | - Chunping Zhang
- Department of Cell Biology, School of Medicine, Nanchang University, Nanchang, China
| |
Collapse
|
14
|
Long-term oncologic outcome and its prognostic indicators in reproductive-age women with ovarian clear-cell carcinoma. Arch Gynecol Obstet 2019; 300:717-724. [PMID: 31165243 DOI: 10.1007/s00404-019-05203-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 05/30/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Clear-cell carcinoma (CCC) in reproductive-age women is likely to become an increasingly critical issue regarding possibilities of infertility, hormonal dysfunction, and mortality. The aim of this study was to examine the long-term oncologic outcome and its prognostic indicators based on a multicentric cohort of young patients with CCC. PATIENTS AND METHODS During the period of 1990-2015, a total of 164 patients aged 45-year-old or younger were enrolled in the study. Clinicopathologic data of these young patients with CCC collected under a centralized pathological review system were subjected to uni- and multivariable analyses to evaluate overall survival (OS). RESULTS The median follow-up was 73.8 months (range 5.2-244.2) in the surviving patients. Among these patients, 104 (63.4%) had FIGO I disease, and 22 (13.4%), 31 (18.9%), and 7 (4.3%) had II, III, and IV disease, respectively. The 5-year OS rate was 74.5%. On stratification by the FIGO stage, the 5-year OS rates were as follows: stage I (90.2%), stage II (57.9%), and stage III/IV (39.3%), respectively (P < 0.0001). Confining analysis to stage I patients, there was no difference in OS between those who underwent fertility-sparing surgery and those who received radical surgery (P = 0.1593). In relapsed patients, the median survival after recurrence was 11.6 months. In multivariable analysis of stage I patients, the capsule status was an independent prognostic indicator of OS {IC2/IC3 vs. IA/IC1: HR 4.293 (95% CI 1.140-16.422), P = 0.0318}. CONCLUSION CCC patients staged greater than IC2/IC3 show a markedly increased risk of mortality. Thus, it is important to diagnose patients staged under IC2/IC3.
Collapse
|
15
|
Yano M, Katoh T, Miyazawa M, Miyazawa M, Ogane N, Miwa M, Hasegawa K, Narahara H, Yasuda M. Clinicopathological correlation of ARID1A status with HDAC6 and its related factors in ovarian clear cell carcinoma. Sci Rep 2019; 9:2397. [PMID: 30787326 PMCID: PMC6382831 DOI: 10.1038/s41598-019-38653-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/10/2018] [Indexed: 12/12/2022] Open
Abstract
Ovarian clear cell carcinoma (OCCC) is associated with a frequent loss in ARID1A function. ARID1A reportedly suppresses histone deacetylase (HDAC)6 in OCCC directly. Here, we evaluated the clinical significance of HDAC6 expression and its related factors in terms of ARID1A status. Immunohistochemical expression of HDAC6, hypoxia inducible factors-1α (HIF-1α), programmed death-1 ligand (PD-L1), CD44 (cancer stem cell marker), and ARID1A was analysed for 106 OCCC patients. High nuclear HDAC6 expression correlated with patient death (p = 0.038). In the multivariate analysis of overall survival, surgical status (complete or incomplete resection) (hazard ratio (HR) = 17.5; p = <0.001), HDAC6 nuclear expression (HR = 1.68; p = 0.034), and PD-L1 expression (HR = 1.95; p = 0.022) were the independent prognostic factors. HDAC6 upregulation and ARID1A loss did not necessarily occur simultaneously. High HDAC6 expression was associated with poor prognosis in OCCC with ARID1A loss; this was not observed without ARID1A loss. HDAC6 expression showed a significant positive correlation with HIF-1α, PD-L1, and CD44. In OCCC, HDAC6 involvement in prognosis depended on ARID1A status. HDAC6 also led to immuno- and hypoxia- tolerance and cancer stem cell phenotype. HDAC6 is a promising therapeutic target for OCCC with loss of ARID1A.
Collapse
Affiliation(s)
- Mitsutake Yano
- Department of Pathology, Saitama Medical University International Medical Centre, Saitama, Japan. .,Departments of Obstetrics and Gynaecology, Oita University Faculty of Medicine, Oita, Japan.
| | - Tomomi Katoh
- Department of Pathology, Saitama Medical University International Medical Centre, Saitama, Japan
| | - Mariko Miyazawa
- Department of Obstetrics and Gynaecology, Tokai University School of Medicine, Kanagawa, Japan
| | - Masaki Miyazawa
- Department of Obstetrics and Gynaecology, Tokai University School of Medicine, Kanagawa, Japan
| | - Naoki Ogane
- Division of Pathology, Ashigarakami Hospital, Kanagawa, Japan
| | - Maiko Miwa
- Department of Gynaecologic Oncology, Saitama Medical University International Medical Centre, Saitama, Japan
| | - Kosei Hasegawa
- Department of Gynaecologic Oncology, Saitama Medical University International Medical Centre, Saitama, Japan
| | - Hisashi Narahara
- Departments of Obstetrics and Gynaecology, Oita University Faculty of Medicine, Oita, Japan
| | - Masanori Yasuda
- Department of Pathology, Saitama Medical University International Medical Centre, Saitama, Japan.
| |
Collapse
|
16
|
Zhang Z, Huang Y, Zhang J, Liu Z, Lin Q, Wang Z. Activation of NF-κB signaling pathway during HCG-induced VEGF expression in luteal cells. Cell Biol Int 2019; 43:344-349. [PMID: 30597662 DOI: 10.1002/cbin.11090] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 12/21/2018] [Indexed: 12/25/2022]
Abstract
Vascular endothelial growth factor (VEGF) plays an essential role in luteal angiogenesis, the present study therefore utilized luteal cells cultured in vitro to further investigate the activation and contribution of nuclear factor (NF)-κB to VEGF expression induced by human chorionic gonadotrophin (HCG). The present results showed HCG induced VEGF expression as well as hypoxia-inducible factor (HIF)-1α mRNA and protein expressions, which was blocked by NF-κB inhibitor pyrrolidine dithiocarbamate (PDTC). Further analysis found that these increases of VEGF and HIF-1α mRNA induced by HCG were also blocked by NF-κB siRNA transfection, which was consistent with PDTC treatment. However, HIF-1α siRNA treatment significantly decreased HCG induced-VEGF expression with no effect on NF-κB mRNA expression. Furthermore, combination of HIF-1α siRNA and PDTC treatment did not further decrease VEGF mRNA expression, and the result of chromatin immunoprecipitation indicated NF-κB may regulate HIF-1α transcription through binding with its promoter. Taken together, the present results clearly demonstrated that NF-κB was activated to regulate VEGF expression by increasing HIF-1α transcription in luteal cells treated with HCG. Therefore, the present study provided a new and important mechanism of luteal angiogenesis during the formation of corpus luteum in mammals.
Collapse
Affiliation(s)
- Zhenghong Zhang
- Provincial Key Laboratory for Developmental Biology and Neurosciences, Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal University, No. 8, Shangsan Road, Fuzhou 350007, P. R. China
| | - Yuxiu Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, P. R. China
| | - Jingwei Zhang
- Provincial Key Laboratory for Developmental Biology and Neurosciences, Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal University, No. 8, Shangsan Road, Fuzhou 350007, P. R. China
| | - Zhaoyuan Liu
- Provincial Key Laboratory for Developmental Biology and Neurosciences, Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal University, No. 8, Shangsan Road, Fuzhou 350007, P. R. China
| | - Qingqiang Lin
- Provincial Key Laboratory for Developmental Biology and Neurosciences, Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal University, No. 8, Shangsan Road, Fuzhou 350007, P. R. China
| | - Zhengchao Wang
- Provincial Key Laboratory for Developmental Biology and Neurosciences, Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal University, No. 8, Shangsan Road, Fuzhou 350007, P. R. China
| |
Collapse
|
17
|
Peng W, Zhang S, Zhang Z, Xu P, Mao D, Huang S, Chen B, Zhang C, Zhang S. Jianpi Jiedu decoction, a traditional Chinese medicine formula, inhibits tumorigenesis, metastasis, and angiogenesis through the mTOR/HIF-1α/VEGF pathway. JOURNAL OF ETHNOPHARMACOLOGY 2018; 224:140-148. [PMID: 29852266 DOI: 10.1016/j.jep.2018.05.039] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/20/2018] [Accepted: 05/26/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese medicine has been utilized for the treatment of cancer. Jianpi Jiedu decoction (JPJD), a traditional Chinese medicine formula, has been used for the treatment of colorectal cancer for decades. However, the underlying molecular mechanistic basis for the effect of JPJD on colorectal cancer is poorly understood. AIM OF THE STUDY The aim of this study was to identify the effects of JPJD on human colon cancer cells in vitro as well as in vivo and to investigate the mechanistic basis for the anticancer effect of JPJD. MATERIALS AND METHODS The in vitro antitumor activity of JPJD was assessed by MTT assay, flow cytometric analysis, wound-healing assay, transwell assays, and tube formation assays in order to assess cell activity, apoptosis, migration, invasion, and angiogenesis, respectively. The anticancer properties of JPJD in vivo were assessed by immunohistochemistry in a nude mouse xenograft model of HCT116 cells. In addition, the level of mTOR/HIF-1α/VEGF signaling pathway proteins in HCT116 cells and tumor tissue was evaluated by immunoblotting. RESULTS In vitro, JPJD significantly inhibited colorectal cancer cell lines viability and proliferation. Flow cytometry analysis demonstrated JPJD to induce HCT116 cell apoptosis. Additionally, JPJD effectively suppressed tumor cell migration, invasion, and angiogenesis by inhibiting the mTOR/HIF-1α/VEGF signaling pathway. In vivo, JPJD significantly inhibited HCT116 tumor growth in athymic nude mice, decreased the levels of CD34 as well as VEGF, and downregulated the mTOR/HIF-1α/VEGF pathway. CONCLUSIONS JPJD treatment produced anti-colorectal tumor effects by inhibiting tumorigenesis, metastasis, as well as angiogenesis through the mTOR/HIF-1α/VEGF pathway. Thus, these results provide a strong rationale for the therapeutic use of JPJD in cancer treatment. Further studies are required to investigate the mechanisms underlying anti-CRC effect of JPJD.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Apoptosis/drug effects
- Cell Line, Tumor
- Cell Movement/drug effects
- Cell Survival/drug effects
- Cells, Cultured
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Human Umbilical Vein Endothelial Cells/drug effects
- Human Umbilical Vein Endothelial Cells/physiology
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Male
- Medicine, Chinese Traditional
- Mice, Inbred BALB C
- Mice, Nude
- Neoplasms/drug therapy
- Neoplasms/metabolism
- Neoplasms/pathology
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Neovascularization, Physiologic/drug effects
- Signal Transduction/drug effects
- TOR Serine-Threonine Kinases/metabolism
- Vascular Endothelial Growth Factor A/metabolism
- Wound Healing/drug effects
Collapse
Affiliation(s)
- Weijun Peng
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Shaofang Zhang
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Zheyu Zhang
- Department of Integrated Traditional Chinese & Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Digestive System Department, Affiliated Hospital of Guilin Medical College, Guilin, Guangxi 541001, China
| | - Panpan Xu
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Dan Mao
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Siqi Huang
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Biyue Chen
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Chunhu Zhang
- Department of Integrated Traditional Chinese & Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Sifang Zhang
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
18
|
Ji JX, Wang YK, Cochrane DR, Huntsman DG. Clear cell carcinomas of the ovary and kidney: clarity through genomics. J Pathol 2018; 244:550-564. [PMID: 29344971 DOI: 10.1002/path.5037] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 01/03/2018] [Accepted: 01/05/2018] [Indexed: 12/19/2022]
Abstract
Clear cell ovarian carcinoma (CCOC) and clear cell renal cell carcinoma (ccRCC) both feature clear cytoplasm, owing to the accumulation of cytoplasmic glycogen. Genomic studies have demonstrated several mutational similarities between these two diseases, including frequent alterations in the chromatin remodelling SWI-SNF and cellular proliferation phosphoinositide 3-kinase-mammalian target of rapamycin pathways, as well as a shared hypoxia-like mRNA expression signature. Although many targeted treatment options have been approved for advanced-stage ccRCC, CCOC patients are still treated with conventional platinum and taxane chemotherapy, to which they are resistant. To determine the extent of similarity between these malignancies, we performed unsupervised clustering of mRNA expression data from these cancers. This review highlights the similarities and differences between these two clear cell carcinomas to facilitate knowledge translation within future research efforts. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jennifer X Ji
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yi Kan Wang
- Department of Molecular Oncology, British Columbia Cancer Agency, BC, Canada
| | - Dawn R Cochrane
- Department of Molecular Oncology, British Columbia Cancer Agency, BC, Canada
| | - David G Huntsman
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Molecular Oncology, British Columbia Cancer Agency, BC, Canada
| |
Collapse
|
19
|
MicroRNA-424 inhibits cell migration, invasion, and epithelial mesenchymal transition by downregulating doublecortin-like kinase 1 in ovarian clear cell carcinoma. Int J Biochem Cell Biol 2017; 85:66-74. [DOI: 10.1016/j.biocel.2017.01.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 01/12/2017] [Accepted: 01/29/2017] [Indexed: 12/12/2022]
|
20
|
Coothankandaswamy V, Cao S, Xu Y, Prasad PD, Singh PK, Reynolds CP, Yang S, Ogura J, Ganapathy V, Bhutia YD. Amino acid transporter SLC6A14 is a novel and effective drug target for pancreatic cancer. Br J Pharmacol 2016; 173:3292-3306. [PMID: 27747870 PMCID: PMC5738662 DOI: 10.1111/bph.13616] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 08/16/2016] [Accepted: 08/17/2016] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE Pancreatic cancer is a solid tumour that is often fatal. Hence, there is an urgent need to identify new drug targets for this disease. Highly proliferating cancer cells have an increased demand for nutrients and, therefore, need to up-regulate selective amino acid transporters. Here, we investigated which amino acid transporters are up-regulated in pancreatic cancer and whether any of these transporters has potential as a drug target for this fatal disease. EXPERIMENTAL APPROACH The expression of amino acid transporters in pancreatic cancer was analysed using publicly available microarray datasets, and the findings with the transporter SLC6A14 were validated by mRNA and protein analysis. The potential of SLC6A14 as a drug target was evaluated using a pharmacological blocker in vitro and in vivo. KEY RESULTS SLC6A14 was up-regulated several fold in patient-derived xenografts, primary tumour tissues and pancreatic cancer cells lines compared to normal pancreatic tissue or normal pancreatic epithelial cells. The magnitude of the up-regulation of SLC6A14 was the highest among the amino acid transporters examined. A pharmacological blocker of SLC6A14, α-methyltryptophan, induced amino acid starvation in pancreatic cancer cells and reduced the growth and proliferation of these cells, both in vitro and in vivo. CONCLUSION AND IMPLICATIONS The salient features of this study are that SLC6A14 is markedly up-regulated in pancreatic cancer and that pharmacological blockade of this transporter interferes with amino acid nutrition and reduces growth and proliferation of pancreatic cancer cells. These findings identify SLC6A14 as a novel druggable target for pancreatic cancer.
Collapse
Affiliation(s)
- V Coothankandaswamy
- Department of Biochemistry and Molecular BiologyAugusta UniversityAugustaGA30912USA
| | - S Cao
- Department of Biochemistry and Molecular BiologyUniversity of GeorgiaAthensGAUSA
| | - Y Xu
- Department of Biochemistry and Molecular BiologyUniversity of GeorgiaAthensGAUSA
| | - P D Prasad
- Department of Biochemistry and Molecular BiologyAugusta UniversityAugustaGA30912USA
| | - P K Singh
- Eppley Institute for Research in Cancer and Allied DiseasesUniversity of Nebraska Medical CenterOmahaNEUSA
| | - C P Reynolds
- Department of Cell Biology and Biochemistry and Cancer CenterTexas Tech University Health Sciences CenterLubbockTX30912USA
| | - S Yang
- Department of Cell Biology and Biochemistry and Cancer CenterTexas Tech University Health Sciences CenterLubbockTX30912USA
| | - J Ogura
- Department of Cell Biology and Biochemistry and Cancer CenterTexas Tech University Health Sciences CenterLubbockTX30912USA
| | - V Ganapathy
- Department of Cell Biology and Biochemistry and Cancer CenterTexas Tech University Health Sciences CenterLubbockTX30912USA
| | - Y D Bhutia
- Department of Cell Biology and Biochemistry and Cancer CenterTexas Tech University Health Sciences CenterLubbockTX30912USA
| |
Collapse
|
21
|
Sugiyama T, Okamoto A, Enomoto T, Hamano T, Aotani E, Terao Y, Suzuki N, Mikami M, Yaegashi N, Kato K, Yoshikawa H, Yokoyama Y, Tanabe H, Nishino K, Nomura H, Kim JW, Kim BG, Pignata S, Alexandre J, Green J, Isonishi S, Terauchi F, Fujiwara K, Aoki D. Randomized Phase III Trial of Irinotecan Plus Cisplatin Compared With Paclitaxel Plus Carboplatin As First-Line Chemotherapy for Ovarian Clear Cell Carcinoma: JGOG3017/GCIG Trial. J Clin Oncol 2016; 34:2881-7. [PMID: 27400948 DOI: 10.1200/jco.2016.66.9010] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Clear cell carcinoma (CCC) is a rare histologic subtype that demonstrates poor outcomes in epithelial ovarian cancer. The Japanese Gynecologic Oncology Group conducted the first randomized phase III, CCC-specific clinical trial that compared irinotecan and cisplatin (CPT-P) with paclitaxel plus carboplatin (TC) in patients with CCC. PATIENTS AND METHODS Six hundred sixty-seven patients with stage I to IV CCC of the ovary were randomly assigned to receive irinotecan 60 mg/m(2) on days 1, 8, and 15 plus cisplatin 60 mg/m(2) on day 1 (CPT-P group) every 4 weeks for six cycles or paclitaxel 175 mg/m(2) plus carboplatin area under the curve 6.0 mg/mL/min on day 1 every 3 weeks for six cycles (TC group). The primary end point was progression-free survival. Secondary end points were overall survival, overall response rate, and adverse events. RESULTS Six hundred nineteen patients were clinically and pathologically eligible for evaluation. With a median follow-up of 44.3 months, 2-year progression-free survival rates were 73.0% in the CPT-P group and 77.6% in TC group (hazard ratio, 1.17; 95% CI, 0.87 to 1.58; P = .85). Two-year overall survival rates were 85.5% with CPT-P and 87.4% with TC (hazard ratio, 1.13; 95% CI, 0.80 to 1.61; one-sided P = .76). Grade 3/4 anorexia, diarrhea, nausea, vomiting, and febrile neutropenia occurred more frequently with CPT-P, whereas grade 3/4 leukopenia, neutropenia, thrombocytopenia, peripheral sensory neuropathy, and joint pain occurred more frequently with TC. CONCLUSION No significant survival benefit was found for CPT-P. Both regimens were well tolerated, but the toxicity profiles differed significantly. Treatment with existing anticancer agents has limitations to improving the prognosis of CCC.
Collapse
Affiliation(s)
- Toru Sugiyama
- Toru Sugiyama, Iwate Medical University, Iwate; Aikou Okamoto, Hiroshi Tanabe, and Seiji Isonishi, The Jikei University School of Medicine; Tetsutaro Hamano, Kitasato University; Yasuhisa Terao, Juntendo University; Mikio Mikami, Tokai University; Hiroyuki Nomura and Daisuke Aoki, Keio University; Fumitoshi Terauchi, Tokyo Medical University, Tokyo; Takayuki Enomoto and Koji Nishino, Niigata University, Niigata; Eriko Aotani, Kanagawa Academy of Science and Technology; Nao Suzuki, St Marianna University, Kanagawa; Nobuo Yaegashi, Tohoku University, Miyagi; Kiyoko Kato, Kyushu University, Fukuoka; Hiroyuki Yoshikawa, University of Tsukuba, Ibaraki; Yoshihito Yokoyama, Hirosaki University, Aomori; Keiichi Fujiwara, Saitama Medical University International Medical Center, Saitama, Japan; Jae-Weon Kim, Seoul National University; Byoung-Gie Kim, Sungkyunkwan University, Seoul, South Korea; Sandro Pignata, Istituto Nazionale Tumori di Napoli, Naples, Italy; Jerome Alexandre, Hôpital Hôtel-Dieu, Montreal, Quebec, Canada; and John Green, University of Liverpool, Liverpool, United Kingdom
| | - Aikou Okamoto
- Toru Sugiyama, Iwate Medical University, Iwate; Aikou Okamoto, Hiroshi Tanabe, and Seiji Isonishi, The Jikei University School of Medicine; Tetsutaro Hamano, Kitasato University; Yasuhisa Terao, Juntendo University; Mikio Mikami, Tokai University; Hiroyuki Nomura and Daisuke Aoki, Keio University; Fumitoshi Terauchi, Tokyo Medical University, Tokyo; Takayuki Enomoto and Koji Nishino, Niigata University, Niigata; Eriko Aotani, Kanagawa Academy of Science and Technology; Nao Suzuki, St Marianna University, Kanagawa; Nobuo Yaegashi, Tohoku University, Miyagi; Kiyoko Kato, Kyushu University, Fukuoka; Hiroyuki Yoshikawa, University of Tsukuba, Ibaraki; Yoshihito Yokoyama, Hirosaki University, Aomori; Keiichi Fujiwara, Saitama Medical University International Medical Center, Saitama, Japan; Jae-Weon Kim, Seoul National University; Byoung-Gie Kim, Sungkyunkwan University, Seoul, South Korea; Sandro Pignata, Istituto Nazionale Tumori di Napoli, Naples, Italy; Jerome Alexandre, Hôpital Hôtel-Dieu, Montreal, Quebec, Canada; and John Green, University of Liverpool, Liverpool, United Kingdom.
| | - Takayuki Enomoto
- Toru Sugiyama, Iwate Medical University, Iwate; Aikou Okamoto, Hiroshi Tanabe, and Seiji Isonishi, The Jikei University School of Medicine; Tetsutaro Hamano, Kitasato University; Yasuhisa Terao, Juntendo University; Mikio Mikami, Tokai University; Hiroyuki Nomura and Daisuke Aoki, Keio University; Fumitoshi Terauchi, Tokyo Medical University, Tokyo; Takayuki Enomoto and Koji Nishino, Niigata University, Niigata; Eriko Aotani, Kanagawa Academy of Science and Technology; Nao Suzuki, St Marianna University, Kanagawa; Nobuo Yaegashi, Tohoku University, Miyagi; Kiyoko Kato, Kyushu University, Fukuoka; Hiroyuki Yoshikawa, University of Tsukuba, Ibaraki; Yoshihito Yokoyama, Hirosaki University, Aomori; Keiichi Fujiwara, Saitama Medical University International Medical Center, Saitama, Japan; Jae-Weon Kim, Seoul National University; Byoung-Gie Kim, Sungkyunkwan University, Seoul, South Korea; Sandro Pignata, Istituto Nazionale Tumori di Napoli, Naples, Italy; Jerome Alexandre, Hôpital Hôtel-Dieu, Montreal, Quebec, Canada; and John Green, University of Liverpool, Liverpool, United Kingdom
| | - Tetsutaro Hamano
- Toru Sugiyama, Iwate Medical University, Iwate; Aikou Okamoto, Hiroshi Tanabe, and Seiji Isonishi, The Jikei University School of Medicine; Tetsutaro Hamano, Kitasato University; Yasuhisa Terao, Juntendo University; Mikio Mikami, Tokai University; Hiroyuki Nomura and Daisuke Aoki, Keio University; Fumitoshi Terauchi, Tokyo Medical University, Tokyo; Takayuki Enomoto and Koji Nishino, Niigata University, Niigata; Eriko Aotani, Kanagawa Academy of Science and Technology; Nao Suzuki, St Marianna University, Kanagawa; Nobuo Yaegashi, Tohoku University, Miyagi; Kiyoko Kato, Kyushu University, Fukuoka; Hiroyuki Yoshikawa, University of Tsukuba, Ibaraki; Yoshihito Yokoyama, Hirosaki University, Aomori; Keiichi Fujiwara, Saitama Medical University International Medical Center, Saitama, Japan; Jae-Weon Kim, Seoul National University; Byoung-Gie Kim, Sungkyunkwan University, Seoul, South Korea; Sandro Pignata, Istituto Nazionale Tumori di Napoli, Naples, Italy; Jerome Alexandre, Hôpital Hôtel-Dieu, Montreal, Quebec, Canada; and John Green, University of Liverpool, Liverpool, United Kingdom
| | - Eriko Aotani
- Toru Sugiyama, Iwate Medical University, Iwate; Aikou Okamoto, Hiroshi Tanabe, and Seiji Isonishi, The Jikei University School of Medicine; Tetsutaro Hamano, Kitasato University; Yasuhisa Terao, Juntendo University; Mikio Mikami, Tokai University; Hiroyuki Nomura and Daisuke Aoki, Keio University; Fumitoshi Terauchi, Tokyo Medical University, Tokyo; Takayuki Enomoto and Koji Nishino, Niigata University, Niigata; Eriko Aotani, Kanagawa Academy of Science and Technology; Nao Suzuki, St Marianna University, Kanagawa; Nobuo Yaegashi, Tohoku University, Miyagi; Kiyoko Kato, Kyushu University, Fukuoka; Hiroyuki Yoshikawa, University of Tsukuba, Ibaraki; Yoshihito Yokoyama, Hirosaki University, Aomori; Keiichi Fujiwara, Saitama Medical University International Medical Center, Saitama, Japan; Jae-Weon Kim, Seoul National University; Byoung-Gie Kim, Sungkyunkwan University, Seoul, South Korea; Sandro Pignata, Istituto Nazionale Tumori di Napoli, Naples, Italy; Jerome Alexandre, Hôpital Hôtel-Dieu, Montreal, Quebec, Canada; and John Green, University of Liverpool, Liverpool, United Kingdom
| | - Yasuhisa Terao
- Toru Sugiyama, Iwate Medical University, Iwate; Aikou Okamoto, Hiroshi Tanabe, and Seiji Isonishi, The Jikei University School of Medicine; Tetsutaro Hamano, Kitasato University; Yasuhisa Terao, Juntendo University; Mikio Mikami, Tokai University; Hiroyuki Nomura and Daisuke Aoki, Keio University; Fumitoshi Terauchi, Tokyo Medical University, Tokyo; Takayuki Enomoto and Koji Nishino, Niigata University, Niigata; Eriko Aotani, Kanagawa Academy of Science and Technology; Nao Suzuki, St Marianna University, Kanagawa; Nobuo Yaegashi, Tohoku University, Miyagi; Kiyoko Kato, Kyushu University, Fukuoka; Hiroyuki Yoshikawa, University of Tsukuba, Ibaraki; Yoshihito Yokoyama, Hirosaki University, Aomori; Keiichi Fujiwara, Saitama Medical University International Medical Center, Saitama, Japan; Jae-Weon Kim, Seoul National University; Byoung-Gie Kim, Sungkyunkwan University, Seoul, South Korea; Sandro Pignata, Istituto Nazionale Tumori di Napoli, Naples, Italy; Jerome Alexandre, Hôpital Hôtel-Dieu, Montreal, Quebec, Canada; and John Green, University of Liverpool, Liverpool, United Kingdom
| | - Nao Suzuki
- Toru Sugiyama, Iwate Medical University, Iwate; Aikou Okamoto, Hiroshi Tanabe, and Seiji Isonishi, The Jikei University School of Medicine; Tetsutaro Hamano, Kitasato University; Yasuhisa Terao, Juntendo University; Mikio Mikami, Tokai University; Hiroyuki Nomura and Daisuke Aoki, Keio University; Fumitoshi Terauchi, Tokyo Medical University, Tokyo; Takayuki Enomoto and Koji Nishino, Niigata University, Niigata; Eriko Aotani, Kanagawa Academy of Science and Technology; Nao Suzuki, St Marianna University, Kanagawa; Nobuo Yaegashi, Tohoku University, Miyagi; Kiyoko Kato, Kyushu University, Fukuoka; Hiroyuki Yoshikawa, University of Tsukuba, Ibaraki; Yoshihito Yokoyama, Hirosaki University, Aomori; Keiichi Fujiwara, Saitama Medical University International Medical Center, Saitama, Japan; Jae-Weon Kim, Seoul National University; Byoung-Gie Kim, Sungkyunkwan University, Seoul, South Korea; Sandro Pignata, Istituto Nazionale Tumori di Napoli, Naples, Italy; Jerome Alexandre, Hôpital Hôtel-Dieu, Montreal, Quebec, Canada; and John Green, University of Liverpool, Liverpool, United Kingdom
| | - Mikio Mikami
- Toru Sugiyama, Iwate Medical University, Iwate; Aikou Okamoto, Hiroshi Tanabe, and Seiji Isonishi, The Jikei University School of Medicine; Tetsutaro Hamano, Kitasato University; Yasuhisa Terao, Juntendo University; Mikio Mikami, Tokai University; Hiroyuki Nomura and Daisuke Aoki, Keio University; Fumitoshi Terauchi, Tokyo Medical University, Tokyo; Takayuki Enomoto and Koji Nishino, Niigata University, Niigata; Eriko Aotani, Kanagawa Academy of Science and Technology; Nao Suzuki, St Marianna University, Kanagawa; Nobuo Yaegashi, Tohoku University, Miyagi; Kiyoko Kato, Kyushu University, Fukuoka; Hiroyuki Yoshikawa, University of Tsukuba, Ibaraki; Yoshihito Yokoyama, Hirosaki University, Aomori; Keiichi Fujiwara, Saitama Medical University International Medical Center, Saitama, Japan; Jae-Weon Kim, Seoul National University; Byoung-Gie Kim, Sungkyunkwan University, Seoul, South Korea; Sandro Pignata, Istituto Nazionale Tumori di Napoli, Naples, Italy; Jerome Alexandre, Hôpital Hôtel-Dieu, Montreal, Quebec, Canada; and John Green, University of Liverpool, Liverpool, United Kingdom
| | - Nobuo Yaegashi
- Toru Sugiyama, Iwate Medical University, Iwate; Aikou Okamoto, Hiroshi Tanabe, and Seiji Isonishi, The Jikei University School of Medicine; Tetsutaro Hamano, Kitasato University; Yasuhisa Terao, Juntendo University; Mikio Mikami, Tokai University; Hiroyuki Nomura and Daisuke Aoki, Keio University; Fumitoshi Terauchi, Tokyo Medical University, Tokyo; Takayuki Enomoto and Koji Nishino, Niigata University, Niigata; Eriko Aotani, Kanagawa Academy of Science and Technology; Nao Suzuki, St Marianna University, Kanagawa; Nobuo Yaegashi, Tohoku University, Miyagi; Kiyoko Kato, Kyushu University, Fukuoka; Hiroyuki Yoshikawa, University of Tsukuba, Ibaraki; Yoshihito Yokoyama, Hirosaki University, Aomori; Keiichi Fujiwara, Saitama Medical University International Medical Center, Saitama, Japan; Jae-Weon Kim, Seoul National University; Byoung-Gie Kim, Sungkyunkwan University, Seoul, South Korea; Sandro Pignata, Istituto Nazionale Tumori di Napoli, Naples, Italy; Jerome Alexandre, Hôpital Hôtel-Dieu, Montreal, Quebec, Canada; and John Green, University of Liverpool, Liverpool, United Kingdom
| | - Kiyoko Kato
- Toru Sugiyama, Iwate Medical University, Iwate; Aikou Okamoto, Hiroshi Tanabe, and Seiji Isonishi, The Jikei University School of Medicine; Tetsutaro Hamano, Kitasato University; Yasuhisa Terao, Juntendo University; Mikio Mikami, Tokai University; Hiroyuki Nomura and Daisuke Aoki, Keio University; Fumitoshi Terauchi, Tokyo Medical University, Tokyo; Takayuki Enomoto and Koji Nishino, Niigata University, Niigata; Eriko Aotani, Kanagawa Academy of Science and Technology; Nao Suzuki, St Marianna University, Kanagawa; Nobuo Yaegashi, Tohoku University, Miyagi; Kiyoko Kato, Kyushu University, Fukuoka; Hiroyuki Yoshikawa, University of Tsukuba, Ibaraki; Yoshihito Yokoyama, Hirosaki University, Aomori; Keiichi Fujiwara, Saitama Medical University International Medical Center, Saitama, Japan; Jae-Weon Kim, Seoul National University; Byoung-Gie Kim, Sungkyunkwan University, Seoul, South Korea; Sandro Pignata, Istituto Nazionale Tumori di Napoli, Naples, Italy; Jerome Alexandre, Hôpital Hôtel-Dieu, Montreal, Quebec, Canada; and John Green, University of Liverpool, Liverpool, United Kingdom
| | - Hiroyuki Yoshikawa
- Toru Sugiyama, Iwate Medical University, Iwate; Aikou Okamoto, Hiroshi Tanabe, and Seiji Isonishi, The Jikei University School of Medicine; Tetsutaro Hamano, Kitasato University; Yasuhisa Terao, Juntendo University; Mikio Mikami, Tokai University; Hiroyuki Nomura and Daisuke Aoki, Keio University; Fumitoshi Terauchi, Tokyo Medical University, Tokyo; Takayuki Enomoto and Koji Nishino, Niigata University, Niigata; Eriko Aotani, Kanagawa Academy of Science and Technology; Nao Suzuki, St Marianna University, Kanagawa; Nobuo Yaegashi, Tohoku University, Miyagi; Kiyoko Kato, Kyushu University, Fukuoka; Hiroyuki Yoshikawa, University of Tsukuba, Ibaraki; Yoshihito Yokoyama, Hirosaki University, Aomori; Keiichi Fujiwara, Saitama Medical University International Medical Center, Saitama, Japan; Jae-Weon Kim, Seoul National University; Byoung-Gie Kim, Sungkyunkwan University, Seoul, South Korea; Sandro Pignata, Istituto Nazionale Tumori di Napoli, Naples, Italy; Jerome Alexandre, Hôpital Hôtel-Dieu, Montreal, Quebec, Canada; and John Green, University of Liverpool, Liverpool, United Kingdom
| | - Yoshihito Yokoyama
- Toru Sugiyama, Iwate Medical University, Iwate; Aikou Okamoto, Hiroshi Tanabe, and Seiji Isonishi, The Jikei University School of Medicine; Tetsutaro Hamano, Kitasato University; Yasuhisa Terao, Juntendo University; Mikio Mikami, Tokai University; Hiroyuki Nomura and Daisuke Aoki, Keio University; Fumitoshi Terauchi, Tokyo Medical University, Tokyo; Takayuki Enomoto and Koji Nishino, Niigata University, Niigata; Eriko Aotani, Kanagawa Academy of Science and Technology; Nao Suzuki, St Marianna University, Kanagawa; Nobuo Yaegashi, Tohoku University, Miyagi; Kiyoko Kato, Kyushu University, Fukuoka; Hiroyuki Yoshikawa, University of Tsukuba, Ibaraki; Yoshihito Yokoyama, Hirosaki University, Aomori; Keiichi Fujiwara, Saitama Medical University International Medical Center, Saitama, Japan; Jae-Weon Kim, Seoul National University; Byoung-Gie Kim, Sungkyunkwan University, Seoul, South Korea; Sandro Pignata, Istituto Nazionale Tumori di Napoli, Naples, Italy; Jerome Alexandre, Hôpital Hôtel-Dieu, Montreal, Quebec, Canada; and John Green, University of Liverpool, Liverpool, United Kingdom
| | - Hiroshi Tanabe
- Toru Sugiyama, Iwate Medical University, Iwate; Aikou Okamoto, Hiroshi Tanabe, and Seiji Isonishi, The Jikei University School of Medicine; Tetsutaro Hamano, Kitasato University; Yasuhisa Terao, Juntendo University; Mikio Mikami, Tokai University; Hiroyuki Nomura and Daisuke Aoki, Keio University; Fumitoshi Terauchi, Tokyo Medical University, Tokyo; Takayuki Enomoto and Koji Nishino, Niigata University, Niigata; Eriko Aotani, Kanagawa Academy of Science and Technology; Nao Suzuki, St Marianna University, Kanagawa; Nobuo Yaegashi, Tohoku University, Miyagi; Kiyoko Kato, Kyushu University, Fukuoka; Hiroyuki Yoshikawa, University of Tsukuba, Ibaraki; Yoshihito Yokoyama, Hirosaki University, Aomori; Keiichi Fujiwara, Saitama Medical University International Medical Center, Saitama, Japan; Jae-Weon Kim, Seoul National University; Byoung-Gie Kim, Sungkyunkwan University, Seoul, South Korea; Sandro Pignata, Istituto Nazionale Tumori di Napoli, Naples, Italy; Jerome Alexandre, Hôpital Hôtel-Dieu, Montreal, Quebec, Canada; and John Green, University of Liverpool, Liverpool, United Kingdom
| | - Koji Nishino
- Toru Sugiyama, Iwate Medical University, Iwate; Aikou Okamoto, Hiroshi Tanabe, and Seiji Isonishi, The Jikei University School of Medicine; Tetsutaro Hamano, Kitasato University; Yasuhisa Terao, Juntendo University; Mikio Mikami, Tokai University; Hiroyuki Nomura and Daisuke Aoki, Keio University; Fumitoshi Terauchi, Tokyo Medical University, Tokyo; Takayuki Enomoto and Koji Nishino, Niigata University, Niigata; Eriko Aotani, Kanagawa Academy of Science and Technology; Nao Suzuki, St Marianna University, Kanagawa; Nobuo Yaegashi, Tohoku University, Miyagi; Kiyoko Kato, Kyushu University, Fukuoka; Hiroyuki Yoshikawa, University of Tsukuba, Ibaraki; Yoshihito Yokoyama, Hirosaki University, Aomori; Keiichi Fujiwara, Saitama Medical University International Medical Center, Saitama, Japan; Jae-Weon Kim, Seoul National University; Byoung-Gie Kim, Sungkyunkwan University, Seoul, South Korea; Sandro Pignata, Istituto Nazionale Tumori di Napoli, Naples, Italy; Jerome Alexandre, Hôpital Hôtel-Dieu, Montreal, Quebec, Canada; and John Green, University of Liverpool, Liverpool, United Kingdom
| | - Hiroyuki Nomura
- Toru Sugiyama, Iwate Medical University, Iwate; Aikou Okamoto, Hiroshi Tanabe, and Seiji Isonishi, The Jikei University School of Medicine; Tetsutaro Hamano, Kitasato University; Yasuhisa Terao, Juntendo University; Mikio Mikami, Tokai University; Hiroyuki Nomura and Daisuke Aoki, Keio University; Fumitoshi Terauchi, Tokyo Medical University, Tokyo; Takayuki Enomoto and Koji Nishino, Niigata University, Niigata; Eriko Aotani, Kanagawa Academy of Science and Technology; Nao Suzuki, St Marianna University, Kanagawa; Nobuo Yaegashi, Tohoku University, Miyagi; Kiyoko Kato, Kyushu University, Fukuoka; Hiroyuki Yoshikawa, University of Tsukuba, Ibaraki; Yoshihito Yokoyama, Hirosaki University, Aomori; Keiichi Fujiwara, Saitama Medical University International Medical Center, Saitama, Japan; Jae-Weon Kim, Seoul National University; Byoung-Gie Kim, Sungkyunkwan University, Seoul, South Korea; Sandro Pignata, Istituto Nazionale Tumori di Napoli, Naples, Italy; Jerome Alexandre, Hôpital Hôtel-Dieu, Montreal, Quebec, Canada; and John Green, University of Liverpool, Liverpool, United Kingdom
| | - Jae-Weon Kim
- Toru Sugiyama, Iwate Medical University, Iwate; Aikou Okamoto, Hiroshi Tanabe, and Seiji Isonishi, The Jikei University School of Medicine; Tetsutaro Hamano, Kitasato University; Yasuhisa Terao, Juntendo University; Mikio Mikami, Tokai University; Hiroyuki Nomura and Daisuke Aoki, Keio University; Fumitoshi Terauchi, Tokyo Medical University, Tokyo; Takayuki Enomoto and Koji Nishino, Niigata University, Niigata; Eriko Aotani, Kanagawa Academy of Science and Technology; Nao Suzuki, St Marianna University, Kanagawa; Nobuo Yaegashi, Tohoku University, Miyagi; Kiyoko Kato, Kyushu University, Fukuoka; Hiroyuki Yoshikawa, University of Tsukuba, Ibaraki; Yoshihito Yokoyama, Hirosaki University, Aomori; Keiichi Fujiwara, Saitama Medical University International Medical Center, Saitama, Japan; Jae-Weon Kim, Seoul National University; Byoung-Gie Kim, Sungkyunkwan University, Seoul, South Korea; Sandro Pignata, Istituto Nazionale Tumori di Napoli, Naples, Italy; Jerome Alexandre, Hôpital Hôtel-Dieu, Montreal, Quebec, Canada; and John Green, University of Liverpool, Liverpool, United Kingdom
| | - Byoung-Gie Kim
- Toru Sugiyama, Iwate Medical University, Iwate; Aikou Okamoto, Hiroshi Tanabe, and Seiji Isonishi, The Jikei University School of Medicine; Tetsutaro Hamano, Kitasato University; Yasuhisa Terao, Juntendo University; Mikio Mikami, Tokai University; Hiroyuki Nomura and Daisuke Aoki, Keio University; Fumitoshi Terauchi, Tokyo Medical University, Tokyo; Takayuki Enomoto and Koji Nishino, Niigata University, Niigata; Eriko Aotani, Kanagawa Academy of Science and Technology; Nao Suzuki, St Marianna University, Kanagawa; Nobuo Yaegashi, Tohoku University, Miyagi; Kiyoko Kato, Kyushu University, Fukuoka; Hiroyuki Yoshikawa, University of Tsukuba, Ibaraki; Yoshihito Yokoyama, Hirosaki University, Aomori; Keiichi Fujiwara, Saitama Medical University International Medical Center, Saitama, Japan; Jae-Weon Kim, Seoul National University; Byoung-Gie Kim, Sungkyunkwan University, Seoul, South Korea; Sandro Pignata, Istituto Nazionale Tumori di Napoli, Naples, Italy; Jerome Alexandre, Hôpital Hôtel-Dieu, Montreal, Quebec, Canada; and John Green, University of Liverpool, Liverpool, United Kingdom
| | - Sandro Pignata
- Toru Sugiyama, Iwate Medical University, Iwate; Aikou Okamoto, Hiroshi Tanabe, and Seiji Isonishi, The Jikei University School of Medicine; Tetsutaro Hamano, Kitasato University; Yasuhisa Terao, Juntendo University; Mikio Mikami, Tokai University; Hiroyuki Nomura and Daisuke Aoki, Keio University; Fumitoshi Terauchi, Tokyo Medical University, Tokyo; Takayuki Enomoto and Koji Nishino, Niigata University, Niigata; Eriko Aotani, Kanagawa Academy of Science and Technology; Nao Suzuki, St Marianna University, Kanagawa; Nobuo Yaegashi, Tohoku University, Miyagi; Kiyoko Kato, Kyushu University, Fukuoka; Hiroyuki Yoshikawa, University of Tsukuba, Ibaraki; Yoshihito Yokoyama, Hirosaki University, Aomori; Keiichi Fujiwara, Saitama Medical University International Medical Center, Saitama, Japan; Jae-Weon Kim, Seoul National University; Byoung-Gie Kim, Sungkyunkwan University, Seoul, South Korea; Sandro Pignata, Istituto Nazionale Tumori di Napoli, Naples, Italy; Jerome Alexandre, Hôpital Hôtel-Dieu, Montreal, Quebec, Canada; and John Green, University of Liverpool, Liverpool, United Kingdom
| | - Jerome Alexandre
- Toru Sugiyama, Iwate Medical University, Iwate; Aikou Okamoto, Hiroshi Tanabe, and Seiji Isonishi, The Jikei University School of Medicine; Tetsutaro Hamano, Kitasato University; Yasuhisa Terao, Juntendo University; Mikio Mikami, Tokai University; Hiroyuki Nomura and Daisuke Aoki, Keio University; Fumitoshi Terauchi, Tokyo Medical University, Tokyo; Takayuki Enomoto and Koji Nishino, Niigata University, Niigata; Eriko Aotani, Kanagawa Academy of Science and Technology; Nao Suzuki, St Marianna University, Kanagawa; Nobuo Yaegashi, Tohoku University, Miyagi; Kiyoko Kato, Kyushu University, Fukuoka; Hiroyuki Yoshikawa, University of Tsukuba, Ibaraki; Yoshihito Yokoyama, Hirosaki University, Aomori; Keiichi Fujiwara, Saitama Medical University International Medical Center, Saitama, Japan; Jae-Weon Kim, Seoul National University; Byoung-Gie Kim, Sungkyunkwan University, Seoul, South Korea; Sandro Pignata, Istituto Nazionale Tumori di Napoli, Naples, Italy; Jerome Alexandre, Hôpital Hôtel-Dieu, Montreal, Quebec, Canada; and John Green, University of Liverpool, Liverpool, United Kingdom
| | - John Green
- Toru Sugiyama, Iwate Medical University, Iwate; Aikou Okamoto, Hiroshi Tanabe, and Seiji Isonishi, The Jikei University School of Medicine; Tetsutaro Hamano, Kitasato University; Yasuhisa Terao, Juntendo University; Mikio Mikami, Tokai University; Hiroyuki Nomura and Daisuke Aoki, Keio University; Fumitoshi Terauchi, Tokyo Medical University, Tokyo; Takayuki Enomoto and Koji Nishino, Niigata University, Niigata; Eriko Aotani, Kanagawa Academy of Science and Technology; Nao Suzuki, St Marianna University, Kanagawa; Nobuo Yaegashi, Tohoku University, Miyagi; Kiyoko Kato, Kyushu University, Fukuoka; Hiroyuki Yoshikawa, University of Tsukuba, Ibaraki; Yoshihito Yokoyama, Hirosaki University, Aomori; Keiichi Fujiwara, Saitama Medical University International Medical Center, Saitama, Japan; Jae-Weon Kim, Seoul National University; Byoung-Gie Kim, Sungkyunkwan University, Seoul, South Korea; Sandro Pignata, Istituto Nazionale Tumori di Napoli, Naples, Italy; Jerome Alexandre, Hôpital Hôtel-Dieu, Montreal, Quebec, Canada; and John Green, University of Liverpool, Liverpool, United Kingdom
| | - Seiji Isonishi
- Toru Sugiyama, Iwate Medical University, Iwate; Aikou Okamoto, Hiroshi Tanabe, and Seiji Isonishi, The Jikei University School of Medicine; Tetsutaro Hamano, Kitasato University; Yasuhisa Terao, Juntendo University; Mikio Mikami, Tokai University; Hiroyuki Nomura and Daisuke Aoki, Keio University; Fumitoshi Terauchi, Tokyo Medical University, Tokyo; Takayuki Enomoto and Koji Nishino, Niigata University, Niigata; Eriko Aotani, Kanagawa Academy of Science and Technology; Nao Suzuki, St Marianna University, Kanagawa; Nobuo Yaegashi, Tohoku University, Miyagi; Kiyoko Kato, Kyushu University, Fukuoka; Hiroyuki Yoshikawa, University of Tsukuba, Ibaraki; Yoshihito Yokoyama, Hirosaki University, Aomori; Keiichi Fujiwara, Saitama Medical University International Medical Center, Saitama, Japan; Jae-Weon Kim, Seoul National University; Byoung-Gie Kim, Sungkyunkwan University, Seoul, South Korea; Sandro Pignata, Istituto Nazionale Tumori di Napoli, Naples, Italy; Jerome Alexandre, Hôpital Hôtel-Dieu, Montreal, Quebec, Canada; and John Green, University of Liverpool, Liverpool, United Kingdom
| | - Fumitoshi Terauchi
- Toru Sugiyama, Iwate Medical University, Iwate; Aikou Okamoto, Hiroshi Tanabe, and Seiji Isonishi, The Jikei University School of Medicine; Tetsutaro Hamano, Kitasato University; Yasuhisa Terao, Juntendo University; Mikio Mikami, Tokai University; Hiroyuki Nomura and Daisuke Aoki, Keio University; Fumitoshi Terauchi, Tokyo Medical University, Tokyo; Takayuki Enomoto and Koji Nishino, Niigata University, Niigata; Eriko Aotani, Kanagawa Academy of Science and Technology; Nao Suzuki, St Marianna University, Kanagawa; Nobuo Yaegashi, Tohoku University, Miyagi; Kiyoko Kato, Kyushu University, Fukuoka; Hiroyuki Yoshikawa, University of Tsukuba, Ibaraki; Yoshihito Yokoyama, Hirosaki University, Aomori; Keiichi Fujiwara, Saitama Medical University International Medical Center, Saitama, Japan; Jae-Weon Kim, Seoul National University; Byoung-Gie Kim, Sungkyunkwan University, Seoul, South Korea; Sandro Pignata, Istituto Nazionale Tumori di Napoli, Naples, Italy; Jerome Alexandre, Hôpital Hôtel-Dieu, Montreal, Quebec, Canada; and John Green, University of Liverpool, Liverpool, United Kingdom
| | - Keiichi Fujiwara
- Toru Sugiyama, Iwate Medical University, Iwate; Aikou Okamoto, Hiroshi Tanabe, and Seiji Isonishi, The Jikei University School of Medicine; Tetsutaro Hamano, Kitasato University; Yasuhisa Terao, Juntendo University; Mikio Mikami, Tokai University; Hiroyuki Nomura and Daisuke Aoki, Keio University; Fumitoshi Terauchi, Tokyo Medical University, Tokyo; Takayuki Enomoto and Koji Nishino, Niigata University, Niigata; Eriko Aotani, Kanagawa Academy of Science and Technology; Nao Suzuki, St Marianna University, Kanagawa; Nobuo Yaegashi, Tohoku University, Miyagi; Kiyoko Kato, Kyushu University, Fukuoka; Hiroyuki Yoshikawa, University of Tsukuba, Ibaraki; Yoshihito Yokoyama, Hirosaki University, Aomori; Keiichi Fujiwara, Saitama Medical University International Medical Center, Saitama, Japan; Jae-Weon Kim, Seoul National University; Byoung-Gie Kim, Sungkyunkwan University, Seoul, South Korea; Sandro Pignata, Istituto Nazionale Tumori di Napoli, Naples, Italy; Jerome Alexandre, Hôpital Hôtel-Dieu, Montreal, Quebec, Canada; and John Green, University of Liverpool, Liverpool, United Kingdom
| | - Daisuke Aoki
- Toru Sugiyama, Iwate Medical University, Iwate; Aikou Okamoto, Hiroshi Tanabe, and Seiji Isonishi, The Jikei University School of Medicine; Tetsutaro Hamano, Kitasato University; Yasuhisa Terao, Juntendo University; Mikio Mikami, Tokai University; Hiroyuki Nomura and Daisuke Aoki, Keio University; Fumitoshi Terauchi, Tokyo Medical University, Tokyo; Takayuki Enomoto and Koji Nishino, Niigata University, Niigata; Eriko Aotani, Kanagawa Academy of Science and Technology; Nao Suzuki, St Marianna University, Kanagawa; Nobuo Yaegashi, Tohoku University, Miyagi; Kiyoko Kato, Kyushu University, Fukuoka; Hiroyuki Yoshikawa, University of Tsukuba, Ibaraki; Yoshihito Yokoyama, Hirosaki University, Aomori; Keiichi Fujiwara, Saitama Medical University International Medical Center, Saitama, Japan; Jae-Weon Kim, Seoul National University; Byoung-Gie Kim, Sungkyunkwan University, Seoul, South Korea; Sandro Pignata, Istituto Nazionale Tumori di Napoli, Naples, Italy; Jerome Alexandre, Hôpital Hôtel-Dieu, Montreal, Quebec, Canada; and John Green, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
22
|
Effect of Target Therapy on the Content of Transcription and Growth Factors, Protein Kinase TOR, and Activity of Intracellular Proteases in Patients with Metastatic Renal Cell Carcinoma. Bull Exp Biol Med 2016; 160:798-801. [PMID: 27165064 DOI: 10.1007/s10517-016-3313-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Indexed: 12/20/2022]
Abstract
We analyzed the dynamics of the expression of transcription factors, VEGF and its receptor VEGFR2, serine-threonine protein kinase mTOR and activity of proteasome and calpain in patients with metastatic renal cancer during therapy with tyrosine kinase inhibitor Votrient and mTOR blocker Afinitor. The expression of hypoxic nuclear factor HIF-1α in the tumor tissue decreased during therapy with the target preparations. The decrease of VEGF and its receptor VEGFR2 was observed only in patients treated with mTOR inhibitor. The increase in calpain activity in the tumor tissue was observed in both groups. These findings extend our understanding of the mechanism of action of target anticancer preparations as allow considering the studied markers as predictors in choosing optimal therapy.
Collapse
|
23
|
Mabuchi S, Sugiyama T, Kimura T. Clear cell carcinoma of the ovary: molecular insights and future therapeutic perspectives. J Gynecol Oncol 2016; 27:e31. [PMID: 27029752 PMCID: PMC4823362 DOI: 10.3802/jgo.2016.27.e31] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 01/12/2016] [Accepted: 01/15/2016] [Indexed: 12/14/2022] Open
Abstract
Clear cell carcinoma (CCC) of the ovary is known to show poorer sensitivity to chemotherapeutic agents and to be associated with a worse prognosis than the more common serous adenocarcinoma or endometrioid adenocarcinoma. To improve the survival of patients with ovarian CCC, the deeper understanding of the mechanism of CCC carcinogenesis as well as the efforts to develop novel treatment strategies in the setting of both front-line treatment and salvage treatment for recurrent disease are needed. In this presentation, we first summarize the mechanism responsible for carcinogenesis. Then, we highlight the promising therapeutic targets in ovarian CCC and provide information on the novel agents which inhibit these molecular targets. Moreover, we discuss on the cytotoxic anti-cancer agents that can be best combined with targeted agents in the treatment of ovarian CCC.
Collapse
Affiliation(s)
- Seiji Mabuchi
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan.
| | - Toru Sugiyama
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, Morioka, Japan
| | - Tadashi Kimura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
24
|
Andorfer P, Heuwieser A, Heinzel A, Lukas A, Mayer B, Perco P. Vascular endothelial growth factor A as predictive marker for mTOR inhibition in relapsing high-grade serous ovarian cancer. BMC SYSTEMS BIOLOGY 2016; 10:33. [PMID: 27090655 PMCID: PMC4836190 DOI: 10.1186/s12918-016-0278-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 04/13/2016] [Indexed: 02/02/2023]
Abstract
Background Development of resistance against first line drug therapy including cisplatin and paclitaxel in high-grade serous ovarian cancer (HGSOC) presents a major challenge. Identifying drug candidates breaking resistance, ideally combined with predictive biomarkers allowing precision use are needed for prolonging progression free survival of ovarian cancer patients. Modeling of molecular processes driving drug resistance in tumor tissue further combined with mechanism of action of drugs provides a strategy for identification of candidate drugs and associated predictive biomarkers. Results Consolidation of transcriptomics profiles and biomedical literature mining results provides 1242 proteins linked with ovarian cancer drug resistance. Integrating this set on a protein interaction network followed by graph segmentation results in a molecular process model representation of drug resistant HGSOC embedding 409 proteins in 24 molecular processes. Utilizing independent transcriptomics profiles with follow-up data on progression free survival allows deriving molecular biomarker-based classifiers for predicting recurrence under first line therapy. Biomarkers of specific relevance are identified in a molecular process encapsulating TGF-beta, mTOR, Jak-STAT and Neurotrophin signaling. Mechanism of action molecular model representations of cisplatin and paclitaxel embed the very same signaling components, and specifically proteins afflicted with the activation status of the mTOR pathway become evident, including VEGFA. Analyzing mechanism of action interference of the mTOR inhibitor sirolimus shows specific impact on the drug resistance signature imposed by cisplatin and paclitaxel, further holding evidence for a synthetic lethal interaction to paclitaxel mechanism of action involving cyclin D1. Conclusions Stratifying drug resistant high grade serous ovarian cancer via VEGFA, and specifically treating with mTOR inhibitors in case of activation of the pathway may allow adding precision for overcoming resistance to first line therapy.
Collapse
Affiliation(s)
- Peter Andorfer
- emergentec biodevelopment GmbH, Gersthofer Strasse 29-31, 1180, Vienna, Austria
| | - Alexander Heuwieser
- emergentec biodevelopment GmbH, Gersthofer Strasse 29-31, 1180, Vienna, Austria
| | - Andreas Heinzel
- emergentec biodevelopment GmbH, Gersthofer Strasse 29-31, 1180, Vienna, Austria
| | - Arno Lukas
- emergentec biodevelopment GmbH, Gersthofer Strasse 29-31, 1180, Vienna, Austria
| | - Bernd Mayer
- emergentec biodevelopment GmbH, Gersthofer Strasse 29-31, 1180, Vienna, Austria
| | - Paul Perco
- emergentec biodevelopment GmbH, Gersthofer Strasse 29-31, 1180, Vienna, Austria.
| |
Collapse
|
25
|
Kurdi A, De Meyer GRY, Martinet W. Potential therapeutic effects of mTOR inhibition in atherosclerosis. Br J Clin Pharmacol 2015; 82:1267-1279. [PMID: 26551391 DOI: 10.1111/bcp.12820] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 11/03/2015] [Accepted: 11/04/2015] [Indexed: 12/13/2022] Open
Abstract
Despite significant improvement in the management of atherosclerosis, this slowly progressing disease continues to affect countless patients around the world. Recently, the mechanistic target of rapamycin (mTOR) has been identified as a pre-eminent factor in the development of atherosclerosis. mTOR is a constitutively active kinase found in two different multiprotein complexes, mTORC1 and mTORC2. Pharmacological interventions with a class of macrolide immunosuppressive drugs, called rapalogs, have shown undeniable evidence of the value of mTORC1 inhibition to prevent the development of atherosclerotic plaques in several animal models. Rapalog-eluting stents have also shown extraordinary results in humans, even though the exact mechanism for this anti-atherosclerotic effect remains elusive. Unfortunately, rapalogs are known to trigger diverse undesirable effects owing to mTORC1 resistance or mTORC2 inhibition. These adverse effects include dyslipidaemia and insulin resistance, both known triggers of atherosclerosis. Several strategies, such as combination therapy with statins and metformin, have been suggested to oppose rapalog-mediated adverse effects. Statins and metformin are known to inhibit mTORC1 indirectly via 5' adenosine monophosphate-activated protein kinase (AMPK) activation and may hold the key to exploit the full potential of mTORC1 inhibition in the treatment of atherosclerosis. Intermittent regimens and dose reduction have also been proposed to improve rapalog's mTORC1 selectivity, thereby reducing mTORC2-related side effects.
Collapse
Affiliation(s)
- Ammar Kurdi
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
26
|
Potential targets for ovarian clear cell carcinoma: a review of updates and future perspectives. Cancer Cell Int 2015; 15:117. [PMID: 26675567 PMCID: PMC4678619 DOI: 10.1186/s12935-015-0267-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 12/01/2015] [Indexed: 12/16/2022] Open
Abstract
Advances in surgical and medical treatments for ovarian cancer have improved prognoses. Platinum drugs in particular are pivotal for the medical treatment of ovarian cancer. However, previous studies have revealed that some histological subtypes, such as clear cell carcinoma, are resistant to medical treatment, including that with platinum drugs. Consequently, the clinical prognosis of advanced clear cell carcinoma is remarkably inferior, primarily because of its chemoresistant behavior. The prevalence of clear cell carcinoma is approximately 5 % in the West, but in Japan, its prevalence is particularly high, at approximately 25 %. Current medical treatments for advanced clear cell carcinoma are difficult to administer, and they have poor efficacy, warranting the development of novel target-based therapies. In this review, we describe medical treatments for clear cell carcinoma and discuss future prospects for therapy. In particular, we focus on the mechanism of platinum resistance in clear cell carcinoma, including the role of annexin A4, one of the most investigated factors of platinum resistance, as well as the mutant genes and overexpressed proteins such as VEGF, PI3K/AKT/mTOR signaling pathway, ARID1A, hepatocyte nuclear factor-1β, ZNF217. We also review targeted molecular therapeutics for epithelial ovarian cancer and discuss their role in clear cell carcinoma treatment. We review the drugs targeting angiogenesis (bevacizumab, sorafenib, and pazopanib), growth factors (gefitinib, erlotinib, lapatinib, trastuzumab, and AMG479), and signaling pathways (temsirolimus, dasatinib, and imatinib), and other drugs (oregovomab, volociximab, and iniparib). This current review summarizes and discusses the clinical significance of these factors in ovarian clear cell carcinoma as well as their potential mechanisms of action. It may provide new integrative understanding for future studies on their exact role in ovarian clear cell carcinoma.
Collapse
|
27
|
Ho CM, Huang CJ, Huang SH, Chang SF, Cheng WF. Demethylation of HIN-1 reverses paclitaxel-resistance of ovarian clear cell carcinoma through the AKT-mTOR signaling pathway. BMC Cancer 2015; 15:789. [PMID: 26497956 PMCID: PMC4619992 DOI: 10.1186/s12885-015-1744-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 10/09/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Methylation of HIN-1 is associated with poor outcomes in patients with ovarian clear cell carcinoma (OCCC), which is regarded to be an aggressive, chemo-resistant histological subtype. This study aimed to evaluate whether 5-aza-2-deoxycytidine (5-aza-2-dC) can reverse methylation of the HIN-1 gene to restore chemo-sensitivity of OCCC and the possible mechanism. METHODS In vitro flow cytometric analysis and evaluation of caspase-3/7 activity of paclitaxel-sensitive and resistant OCCC cell lines were performed. Methylation status and expression changes of HIN-1 in the OCCC cell lines treated with 5-aza-2-dC were evaluated, and immunohistochemical staining of HIN-1 in OCCC tissues was performed. In vivo tumor growth with or without 5-aza-2-dC treatment was analyzed, and Western blotting of AKT-mTOR signaling-related molecules was performed. RESULTS G2-M phase arrest was absent in paclitaxel-resistant OCCC cells after treatment with the cytotoxic drug. The caspase activities of the chemo-resistant OCCC cells were lower than those of the chemo-sensitive OCCC cells when treated with paclitaxel. Methylation of HIN-1 was noted in paclitaxel-resistant OCCC cell lines and cancerous tissues. 5-aza-2-dC reversed the methylation of HIN-1, re-activated the expression of HIN-1, and then suppressed the in vivo tumor growth of paclitaxel-resistant OCCC cells. Immunoblotting revealed that phospho-AKT473 and phospho-mTOR were significantly increased in HIN-1-methylated paclitaxel-resistant OCCC cell lines. However, the expressions of phospho-AKT at Ser473 and Thr308 and phospho-mTOR decreased in the OCCC cells with a high expression of HIN-1. CONCLUSIONS Demethylating agents can restore the HIN-1 expression in paclitaxel-resistant OCCC cells through the HIN-1-AKT-mTOR signaling pathway to inhibit tumor growth.
Collapse
Affiliation(s)
- Chih-Ming Ho
- Gynecologic Cancer Center, Department of Obstetrics and Gynecology, Cathay General Hospital, Taipei, Taiwan.
- School of Medicine, Fu Jen Catholic University, Hsinchuang, New Taipei City, Taiwan.
- School of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Chi-Jung Huang
- Department of Medical Research, Cathay General Hospital, Sijhih, New Taipei, Taiwan.
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan.
| | - Shih-Hung Huang
- Department of Pathology, Cathay General Hospital, Taipei, Taiwan.
| | - Shwu-Fen Chang
- Graduate Institute of Medical Sciences, School of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Wen-Fang Cheng
- Department of Obstetrics and Gynecology, National Taiwan, University Hospital, Taipei, Taiwan.
- Graduate Institute of Oncology, National Taiwan, University Hospital, Taipei, Taiwan.
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan, University, Taipei, Taiwan.
| |
Collapse
|
28
|
Wu L, Zhang Z, Pan X, Wang Z. Expression and contribution of the HIF-1α/VEGF signaling pathway to luteal development and function in pregnant rats. Mol Med Rep 2015; 12:7153-9. [PMID: 26323652 DOI: 10.3892/mmr.2015.4268] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 08/04/2015] [Indexed: 11/06/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) is vital in normal and abnormal angiogenesis in the ovary, particularly during the early development of the corpus luteum in the ovary. However, the molecular regulation of the expression VEGF during luteal development in vivo remains to be fully elucidated. As the expression of VEGF is mediated by hypoxia‑inducible factor (HIF)‑1α in luteal cells cultured in vitro, determined in our previous study, the present study was performed to confirm the hypothesis that HIF‑1α is induced and then regulates the expression of VEGF and VEGF‑dependent luteal development/function in vivo. This was investigated using a pregnant rat model treated with a small‑molecule inhibitor of HIF‑1α, echinomycin (Ech). The development of the corpus luteum in the pregnant rat ovary was identified via performing assays of the serum progesterone, testosterone and estradiol concentrations by radioimmunoassay, accompanied with determination of the changes in the expression levels of HIF‑1α and VEGF by reverse transcription‑quantitative polymerase chain reaction at different days of the developmental process. On day 5, serum progesterone levels were markedly increased, whereas serum levels of testosterone and estradiol did not change significantly. On day 17, the highest level of serum progesterone was observed, however, this was not the case for testosterone and estradiol. Further analysis of the expression levels of HIF‑1α and VEGF revealed that their changes were consistent with the changes in serum levels of progesterone, which occurred in the development of the corpus luteum in the ovaries of pregnant rats. Further investigation demonstrated that Ech inhibited luteal development through inhibiting the expression of VEGF, mediated by HIF‑1α, and subsequent luteal function, which was determined by detecting changes in serum progesterone on days 8 and 14. Taken together, these results demonstrated that HIF‑1α‑mediated expression of VEGF may be one of the important mechanisms regulating ovarian luteal development in mammals in vivo, which may provide novel strategies in treatment for fertility control and for certain types of ovarian dysfunction, including polycystic ovarian syndrome, ovarian hyperstimulation syndrome and ovarian neoplasia.
Collapse
Affiliation(s)
- Lixiang Wu
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian 350108, P.R. China
| | - Zhenghong Zhang
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian 350108, P.R. China
| | - Xiaoyan Pan
- Department of Histology and Embryology, Jilin Medical College, Jilin, Jilin 132013, P.R. China
| | - Zhengchao Wang
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian 350108, P.R. China
| |
Collapse
|
29
|
Zhang Z, Pang X, Tang Z, Yin D, Wang Z. Overexpression of hypoxia-inducible factor prolyl hydoxylase-2 attenuates hypoxia-induced vascular endothelial growth factor expression in luteal cells. Mol Med Rep 2015; 12:3809-3814. [PMID: 25975603 DOI: 10.3892/mmr.2015.3788] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 04/15/2015] [Indexed: 11/06/2022] Open
Abstract
Vascular endothelial growth factor (VEGF)-dependent angiogenesis has a crucial role in the corpus luteum formation and their functional maintenances in mammalian ovaries. A previous study by our group reported that activation of hypoxia‑inducible factor (HIF)‑1α signaling contributes to the regulation of VEGF expression in the luteal cells (LCs) in response to hypoxia and human chorionic gonadotropin. The present study was designed to test the hypothesis that HIF prolyl‑hydroxylases (PHDs) are expressed in LCs and overexpression of PHD2 attenuates the expression of VEGF induced by hypoxia in LCs. PHD2-overexpressing plasmid was transfected into LC2 cells, and successful plasmid transfection and expression was confirmed by reverse transcription quantitative polymerase chain reaction and western blot analysis. In addition, the present study investigated changes of HIF‑1α and VEGF expression after incubation under hypoxic conditions and PHD2 transfection. PHD2 expression was significantly higher expressed than the other two PHD isoforms, indicating its major role in LCs. Moreover, a significant increase of VEGF mRNA expression was identified after incubation under hypoxic conditions, which was, however, attenuated by PHD2 overexpression in LCs. Further analysis also indicated that this hypoxia‑induced increase in the mRNA expression of VEGF was consistent with increases in the protein levels of HIF‑1α, which is regulated by PHD-mediated degradation. In conclusion, the results of the present study indicated that PHD2 is the main PHD expressed in LCs and hypoxia‑induced VEGF expression can be attenuated by PHD2 overexpression through HIF‑1α‑mediated mechanisms in LCs. This PHD2-mediated transcriptional activation may be one of the mechanisms regulating VEGF expression in LCs during mammalian corpus luteum development.
Collapse
Affiliation(s)
- Zhenghong Zhang
- Provincial Key Laboratory for Developmental Biology and Neurobiology, College of Life Science, Fujian Normal University, Fuzhou, Fujian 350007, P.R. China
| | - Xunsheng Pang
- Provincial Key Laboratory for Developmental Biology and Neurobiology, College of Life Science, Fujian Normal University, Fuzhou, Fujian 350007, P.R. China
| | - Zonghao Tang
- Provincial Key Laboratory for Developmental Biology and Neurobiology, College of Life Science, Fujian Normal University, Fuzhou, Fujian 350007, P.R. China
| | - Dingzhong Yin
- Provincial Key Laboratory for Developmental Biology and Neurobiology, College of Life Science, Fujian Normal University, Fuzhou, Fujian 350007, P.R. China
| | - Zhengchao Wang
- Provincial Key Laboratory for Developmental Biology and Neurobiology, College of Life Science, Fujian Normal University, Fuzhou, Fujian 350007, P.R. China
| |
Collapse
|
30
|
Interplay between receptor tyrosine kinases and hypoxia signaling in cancer. Int J Biochem Cell Biol 2015; 62:101-14. [DOI: 10.1016/j.biocel.2015.02.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 02/24/2015] [Accepted: 02/25/2015] [Indexed: 02/06/2023]
|
31
|
Naruse T, Yanamoto S, Yamada SI, Rokutanda S, Kawakita A, Kawasaki G, Umeda M. Anti-Tumor Effect of the Mammalian Target of Rapamycin Inhibitor Everolimus in Oral Squamous Cell Carcinoma. Pathol Oncol Res 2015; 21:765-73. [DOI: 10.1007/s12253-014-9888-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 12/22/2014] [Indexed: 12/29/2022]
|
32
|
Kim KM, Heo DR, Lee J, Park JS, Baek MG, Yi JM, Kim H, Bang OS. 5,3'-Dihydroxy-6,7,4'-trimethoxyflavanone exerts its anticancer and antiangiogenesis effects through regulation of the Akt/mTOR signaling pathway in human lung cancer cells. Chem Biol Interact 2014; 225:32-9. [PMID: 25446852 DOI: 10.1016/j.cbi.2014.10.033] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 09/26/2014] [Accepted: 10/27/2014] [Indexed: 12/21/2022]
Abstract
5,3'-Dihydroxy-6,7,4'-trimethoxyflavanone (DHTMF) is one of the constituents of Vitex rotundifolia, a medicinal herb that is used for the treatment of various disorders in China and Korea. In this study we evaluated the antitumor and antiangiogeneic activities of DHTMF. DHTMF significantly suppressed growth and induced apoptosis in lung carcinoma cells in a dose-dependent manner, as indicated by a decrease in Bcl-2 levels and increases in Bax and cleaved caspase-3 levels. In addition, DHTMF treatment significantly reduced the phosphorylation of Akt and mammalian target of rapamycin (mTOR), accompanied by reductions in the protein level of hypoxia-inducible factor (HIF-1α) and vascular endothelial growth factor (VEGF), which are key angiogenic molecules in H522 lung cancer cells. Furthermore DHTMF inhibited VEGF-induced angiogenesis, as indicated by reduced expression of CD34, tube formation and migration in human umbilical vein endothelial cells (HUVECs), as well as reduced neovascularization in an in vivo mouse Matrigel plug assay. DHTMF also inhibited phosphorylation of Akt, mTOR, and p70S6K in HUVECs and lung cancer cells. Taken together, our finding indicated that DHTMF inhibits Akt/mTOR signaling and reduces the expression of HIF-1 α and VEGF in tumor cells, which in turns inhibits endothelial cell-mediated angiogenesis. These results suggest that DHTMF inhibits angiogenesis as well as induces apoptosis via the Akt/mTOR pathway and might elicit pharmacological effects that are useful for treatment of lung cancer.
Collapse
Affiliation(s)
- Ki Mo Kim
- Korean Medicine (KM)-Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseong-daero, Yuseong-gu, Daejeon 305-811, Republic of Korea
| | - Deok Rim Heo
- Korean Medicine (KM)-Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseong-daero, Yuseong-gu, Daejeon 305-811, Republic of Korea
| | - Jun Lee
- Korean Medicine (KM)-Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseong-daero, Yuseong-gu, Daejeon 305-811, Republic of Korea
| | - Jong-Shik Park
- Korean Medicine (KM)-Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseong-daero, Yuseong-gu, Daejeon 305-811, Republic of Korea
| | - Myung-Gi Baek
- Korean Medicine (KM)-Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseong-daero, Yuseong-gu, Daejeon 305-811, Republic of Korea
| | - Jin-Mu Yi
- Korean Medicine (KM)-Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseong-daero, Yuseong-gu, Daejeon 305-811, Republic of Korea
| | - Haejin Kim
- Korean Medicine (KM)-Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseong-daero, Yuseong-gu, Daejeon 305-811, Republic of Korea
| | - Ok-Sun Bang
- Korean Medicine (KM)-Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseong-daero, Yuseong-gu, Daejeon 305-811, Republic of Korea.
| |
Collapse
|
33
|
Khemapech N, Pitchaiprasert S, Triratanachat S. Prevalence and clinical significance of mammalian target of rapamycin phosphorylation (p-mTOR) and vascular endothelial growth factor (VEGF) in clear cell carcinoma of the ovary. Asian Pac J Cancer Prev 2014; 13:6357-62. [PMID: 23464458 DOI: 10.7314/apjcp.2012.13.12.6357] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To determine the prevalence of mammalian target of rapamycin phosphorylation (p-mTOR) and vascular endothelial growth factor (VEGF) and any correlation with clinical characteristics and prognosis in ovarian clear cell carcinoma patients. MATERIALS AND METHOD Seventy four paraffin-embedded specimens of such carcinomas frompatients who underwent surgery, received adjuvant chemotherapy and were followed up at King Chulalongkorn Memorial Hospital during January 2002 to December 2008 were stained with rabbit monoclonal IgG p-mTOR and rabbit polyclonal IgG VEGF using immunohistochemical methods. Medical records were reviewed and clinical variables were analysed. RESULTS The prevalence of positive p-mTOR in ovarian clear cell carcinoma was 87.9% and significantly higher in advance-stage than early-stage patients (100% versus 83.6%, P<0.05). Two-year disease free survival and 2-year overall survival in patients with positive p-mTOR expression were 60% and 69.2% with no differences from patients with negative p-mTOR expression (p>0.05). The prevalence of VEGF expression was 63.5% and significantly higher in chemo-sensitive than chemo-resistant patients (70.7% versus 37.5%, P<0.05). Two-year disease free survival and 2-year overall survival in patients with VEGF expression were 72.3% and 83% respectively which were significantly different from patients with negative VEGF expression (p<0.05 ). CONCLUSIONS p-mTOR expression in ovarian clear cell carcinoma was significantly correlated with the stage of disease. VEGF expression was significantly correlated with chemosensitivity, and survival. Further studies of related targeted therapy might be promising.
Collapse
Affiliation(s)
- Nipon Khemapech
- Gynecologic Oncology Division, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| | | | | |
Collapse
|
34
|
δ-Tocotrienol oxazine derivative antagonizes mammary tumor cell compensatory response to CoCl2-induced hypoxia. BIOMED RESEARCH INTERNATIONAL 2014; 2014:285752. [PMID: 25140303 PMCID: PMC4129965 DOI: 10.1155/2014/285752] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 07/03/2014] [Accepted: 07/08/2014] [Indexed: 01/23/2023]
Abstract
In response to low oxygen supply, cancer cells elevate production of HIF-1α, a hypoxia-inducible transcription factor that subsequently acts to stimulate blood vessel formation and promote survival. Studies were conducted to determine the role of δ-tocotrienol and a semisynthetic δ-tocotrienol oxazine derivative, compound 44, on +SA mammary tumor cell hypoxic response. Treatment with 150 µM CoCl2 induced a hypoxic response in +SA mammary tumor cells as evidenced by a large increase in HIF-1α levels, and combined treatment with compound 44 attenuated this response. CoCl2-induced hypoxia was also associated with a large increase in Akt/mTOR signaling, activation of downstream targets p70S6K and eIF-4E1, and a significant increase in VEGF production, and combined treatment with compound 44 blocked this response. Additional in vivo studies showed that intralesional treatment with compound 44 in BALB/c mice bearing +SA mammary tumors significantly decreased the levels of HIF-1α, and this effect was associated with a corresponding decrease in Akt/mTOR signaling and activation of downstream targets p70S6kinase and eIF-4E1. These findings demonstrate that treatment with the δ-tocotrienol oxazine derivative, compound 44, significantly attenuates +SA mammary tumor cell compensatory responses to hypoxia and suggests that this compound may provide benefit in the treatment of rapidly growing solid breast tumors.
Collapse
|
35
|
Abstract
OBJECTIVES Despite improvements in the management of ovarian cancer patients over the last 30 years, there has been only a minimal improvement in overall survival. While targeted therapeutic approaches for the treatment of cancer have evolved, major challenges in ovarian cancer research persist, including the identification of predictive biomarkers with clinical relevance, so that empirical drug selection can be avoided. In this article, we review published genomic analysis studies including data generated in our laboratory and how they have been incorporated into modern clinical trials in a rational and effective way. METHODS Multiple published genomic analysis studies were collected for review and discussion with emphasis on their potential clinical applicability. RESULTS Genomic analysis has been shown to be a powerful tool to identify dysregulated genes, aberrantly activated pathways and to uncover uniqueness of subclasses of ovarian tumors. The application of this technology has provided a solid molecular basis for different clinical behaviors associated with tumor histology and grade. Genomic signatures have been obtained to predict clinical end points for patients with cancer, including response rates, progression-free survival, and overall survival. In addition, genomic analysis has provided opportunities to identify biomarkers, which either result in a modification of existing clinical management or to stratification of patients to novel therapeutic approaches designed as clinical trials. CONCLUSIONS Genomic analyses have accelerated the identification of relevant biomarkers and extended our understanding of the molecular biology of ovarian cancer. This in turn, will hopefully lead to a paradigm shift from empirical, uniform treatment to a more rational, personalized treatment of ovarian cancers. However, validation of potential biomarkers on both the statistical and biological levels is needed to confirm they are of clinical relevance, in order to increase the likelihood that the desired outcome can be predicted and achieved.
Collapse
Affiliation(s)
- W Wei
- Center for Cancer Research, Harvard Medical School
| | | | | | | |
Collapse
|
36
|
Oishi T, Itamochi H, Kudoh A, Nonaka M, Kato M, Nishimura M, Oumi N, Sato S, Naniwa J, Sato S, Shimada M, Kigawa J, Harada T. The PI3K/mTOR dual inhibitor NVP-BEZ235 reduces the growth of ovarian clear cell carcinoma. Oncol Rep 2014; 32:553-8. [PMID: 24927217 DOI: 10.3892/or.2014.3268] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 04/08/2014] [Indexed: 11/06/2022] Open
Abstract
Patients with clear cell carcinoma of the ovary (OCCC) have poor survival due to resistance to standard chemotherapy. OCCC has frequent activating mutations of the PIK3CA gene. The present study was conducted to clarify the efficacy of the inhibition of the PI3K-AKT-mTOR pathway in OCCC. We used 8 OCCC cell lines and 5 ovarian serous adenocarcinoma (OSAC) cell lines. The mutation status of the PIK3CA and KRAS genes was examined by direct sequencing. The IC50 values of NVP-BEZ235 (BEZ235) and temsirolimus were determined by WST-8 assay. Protein expression levels of PI3K-AKT-mTOR pathway molecules were examined by western blotting. Cell cycle distribution was analyzed by flow cytometry. Annexin V staining was used for detecting apoptosis. We also investigated the effects of BEZ235 on OCCC tumor growth in a nude mouse xenograft model. Four of the 8 OCCC cell lines showed a PIK3CA mutation while none of the 5 OSAC cell lines showed a mutation. The IC50 values of BEZ235 for the OCCC cell lines were lower than these values for the OSAC cell lines. The IC50 value of temsirolimus was higher than BEZ235 in the OCCC cell lines. The PIK3CA mutation was more frequently noted in OCCC than OSAC cells, but the sensitivity of these cell lines to BEZ235 or temsirolimus was not related to the mutation status. pHER3 and pAkt proteins were expressed more frequently in OCCC compared with OSAC. However, protein expression levels were distributed widely, and were not related to the sensitivity. Treatment with BEZ235 suppressed expression of pAkt, although treatment with temsirolimus did not. OCCC cells exhibited G1 phase arrest after treatment with BEZ235 and apoptosis with a higher concentration of the agent. BEZ235 significantly inhibited tumor growth in mice bearing OVISE and TU-OC-1 cell tumors. The present study indicated that the PI3K-AKT-mTOR pathway is a potential target for OCCC, and that BEZ235 warrants investigation as a therapeutic agent.
Collapse
Affiliation(s)
- Tetsuro Oishi
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, Yonago, Tottori 683-8504, Japan
| | - Hiroaki Itamochi
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, Yonago, Tottori 683-8504, Japan
| | - Akiko Kudoh
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, Yonago, Tottori 683-8504, Japan
| | - Michiko Nonaka
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, Yonago, Tottori 683-8504, Japan
| | - Misaki Kato
- Tottori University Hospital Cancer Center, Yonago, Tottori 683-8504, Japan
| | - Mayumi Nishimura
- Tottori University Hospital Cancer Center, Yonago, Tottori 683-8504, Japan
| | - Nao Oumi
- Tottori University Hospital Cancer Center, Yonago, Tottori 683-8504, Japan
| | - Seiya Sato
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, Yonago, Tottori 683-8504, Japan
| | - Jun Naniwa
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, Yonago, Tottori 683-8504, Japan
| | - Shinya Sato
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, Yonago, Tottori 683-8504, Japan
| | - Muneaki Shimada
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, Yonago, Tottori 683-8504, Japan
| | - Junzo Kigawa
- Tottori University Hospital Cancer Center, Yonago, Tottori 683-8504, Japan
| | - Tasuku Harada
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, Yonago, Tottori 683-8504, Japan
| |
Collapse
|
37
|
Unwith S, Zhao H, Hennah L, Ma D. The potential role of HIF on tumour progression and dissemination. Int J Cancer 2014; 136:2491-503. [PMID: 24729302 DOI: 10.1002/ijc.28889] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 03/07/2014] [Accepted: 04/03/2014] [Indexed: 12/20/2022]
Abstract
Cancer is the second cause of mortality worldwide, primarily owing to failure to cure metastatic disease. The need to target the metastatic process to reduce mortality is clear and research over the past decade has shown hypoxia-inducible factor-1 (HIF-1) to be one of the promising targets. In order for metastatic disease to be established, multiple steps need to be taken whereby the tumour cells escape into the bloodstream and survive, disseminate and then establish at a premetastatic niche. HIF-1 mediates hypoxia-induced proangiogenic factors such as vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF), which promote extravasation and chemotaxis. The migration of tumour cells is mediated by loss of E-cadherin, which results in a more invasive phenotype; dissemination of the tumour cells by increased vascular permeability and survival in the bloodstream through resistance to apoptosis as well as adhesion at the premetastatic niche are all controlled by factors under the influence of HIF-1. The overexpression of HIF in many aggressive cancer types as well as its role in the establishment of metastatic disease and treatment resistance demonstrate its potential target in therapeutics. Taken together, the role of HIF-1 in cancer and metastatic disease is clear and the need for better treatment targeting metastases is paramount; more aggressive phenotypes with less response to treatment are associated with HIF-1 expression. Our research has shown promise but many questions still remain to be answered.
Collapse
Affiliation(s)
- Sandeep Unwith
- Section of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and, Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, United Kingdom
| | | | | | | |
Collapse
|
38
|
Fujiwara K, Yoshida H, Hasegawa K. Update on nonserous ovarian cancer trials. Ann Oncol 2013; 24 Suppl 10:x46-x47. [DOI: 10.1093/annonc/mdt470] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
39
|
Inhibition of protein kinase C delta attenuates allergic airway inflammation through suppression of PI3K/Akt/mTOR/HIF-1 alpha/VEGF pathway. PLoS One 2013; 8:e81773. [PMID: 24312355 PMCID: PMC3843701 DOI: 10.1371/journal.pone.0081773] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 10/16/2013] [Indexed: 01/05/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) is supposed to contribute to the pathogenesis of allergic airway disease. VEGF expression is regulated by a variety of stimuli such as nitric oxide, growth factors, and hypoxia-inducible factor-1 alpha (HIF-1α). Recently, inhibition of the mammalian target of rapamycin (mTOR) has been shown to alleviate cardinal asthmatic features, including airway hyperresponsiveness, eosinophilic inflammation, and increased vascular permeability in asthma models. Based on these observations, we have investigated whether mTOR is associated with HIF-1α-mediated VEGF expression in allergic asthma. In studies with the mTOR inhibitor rapamycin, we have elucidated the stimulatory role of a mTOR-HIF-1α-VEGF axis in allergic response. Next, the mechanisms by which mTOR is activated to modulate this response have been evaluated. mTOR is known to be regulated by phosphoinositide 3-kinase (PI3K)/Akt or protein kinase C-delta (PKC δ) in various cell types. Consistent with these, our results have revealed that suppression of PKC δ by rottlerin leads to the inhibition of PI3K/Akt activity and the subsequent blockade of a mTOR-HIF-1α-VEGF module, thereby attenuating typical asthmatic attack in a murine model. Thus, the present data indicate that PKC δ is necessary for the modulation of the PI3K/Akt/mTOR signaling cascade, resulting in a tight regulation of HIF-1α activity and VEGF expression. In conclusion, PKC δ may represent a valuable target for innovative therapeutic treatment of allergic airway disease.
Collapse
|
40
|
Alterations of Hypoxia-Induced Factor Signaling Pathway Due to Mammalian Target of Rapamycin (mTOR) Suppression in Ovarian Clear Cell Adenocarcinoma: In Vivo and in Vitro Explorations for Clinical Trial. Int J Gynecol Cancer 2013; 23:1210-8. [DOI: 10.1097/igc.0b013e31829d2d51] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
ObjectivesBefore setting into the clinical trial using a combination of mammalian target of rapamycin (mTOR) inhibitors (rapamycin and everolimus) and other anticancer drugs, this study was conducted to confirm the efficacy of the new therapeutic strategy for ovarian clear cell adenocarcinoma (CCA), which targeted mTOR–hypoxia-induced factor (HIF) signal transduction system.Materials and MethodsUsing the cultured cells of CCA and animal models, alteration of mTOR-HIF cofactors and cell proliferation under the mTOR inhibitor–treated condition were analyzed.ResultsMammalian target of rapamycin–HIF cofactors were inhibited dependent on concentration by mTOR inhibitor, resulting in suppression of the cultured CCA proliferation. However, von Hippel-Lindau was up-regulated at the messenger RNA level. In the nude mice with subcutaneously implanted CCA cells, apoptosis and necrosis were detected especially around the center of the tumors in the mTOR inhibitor–treated group more conspicuously than in the nontreated group. In the assessment of combination therapy with other antitumor agents, a combined treatment with mTOR inhibitor and chemotherapeutic agents caused a significant decrease in tumor size compared to the chemotherapeutic agents–only group.ConclusionsTreatment by mTOR inhibitor is expected to down-regulate the cell proliferation of the CCA as a new therapeutic strategy.
Collapse
|
41
|
Changing Concepts of Hormone Receptor–Positive Advanced Breast Cancer Therapy. Clin Breast Cancer 2013; 13:159-66. [DOI: 10.1016/j.clbc.2012.11.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Revised: 10/22/2012] [Accepted: 11/08/2012] [Indexed: 11/19/2022]
|
42
|
Stathmin Regulates Hypoxia-Inducible Factor-1α Expression through the Mammalian Target of Rapamycin Pathway in Ovarian Clear Cell Adenocarcinoma. ISRN PHARMACOLOGY 2013; 2013:279593. [PMID: 23819061 PMCID: PMC3683482 DOI: 10.1155/2013/279593] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 05/13/2013] [Indexed: 01/17/2023]
Abstract
Stathmin, a microtubule-destabilizing phosphoprotein, is highly expressed in ovarian cancer, but the pathophysiological significance of this protein in ovarian carcinoma cells remains poorly understood. This study reports the involvement of stathmin in the mTOR/HIF-1α/VEGF pathway in ovarian clear cell adenocarcinoma (CCA) during hypoxia. HIF-1α protein and VEGF mRNA levels were markedly elevated in RMG-1 cells, a CCA cell line, cultured under hypoxic conditions. Rapamycin, an inhibitor of mTOR complex 1, reduced the level of HIF-1α and blocked phosphorylation of ribosomal protein S6 kinase 1 (S6K), a transcriptional regulator of mTOR, demonstrating that hypoxia activates mTOR/S6K/HIF-1α signaling in CCA. Furthermore, stathmin knockdown inhibited hypoxia-induced HIF-1α and VEGF expression and S6K phosphorylation. The silencing of stathmin expression also reduced Akt phosphorylation, a critical event in the mTOR/HIF-1α/VEGF signaling pathway. By contrast, stathmin overexpression upregulated hypoxia-induced HIF-1α and VEGF expression in OVCAR-3 cells, another CCA cell line. In addition, suppression of Akt activation by wortmannin, a phosphoinositide 3-kinase (PI3K) inhibitor, decreased HIF-1α and VEGF expression. These results illustrate that regulation of HIF-1α through the PI3K/Akt/mTOR pathway is controlled by stathmin in CCA. Our findings point to a new mechanism of stathmin regulation during ovarian cancer.
Collapse
|
43
|
Sun G, Hu W, Lu Y, Wang Y. A meta-analysis of HIF-1α and esophageal squamous cell carcinoma (ESCC) risk. Pathol Oncol Res 2013; 19:685-93. [PMID: 23658132 DOI: 10.1007/s12253-013-9631-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Accepted: 03/27/2013] [Indexed: 12/18/2022]
Abstract
To investigate the correlation of hypoxia-inducible factor-1α (HIF-1α) expression with clinical prognosis and efficacy of radiochemotherapy in esophageal squamous cell carcinoma (ESCC). Studies assessing the clinical or prognostic significance of HIF-1α expression in ESCC published prior to December 2011 were selected by searching PubMed, EMBASE, Cochrane Library, and (China National Knowledge Infrastructure) CNKI. A meta-analysis was performed to clarify the impact of HIF-1α expression on clinicopathological parameters or survival in ESCC. A total of 16 studies met the inclusion criteria, which included 1261 patients with ESCC. Accordingly, the level of HIF-1α expression in esophageal tissues of patients with ESCC was significantly higher than that in normal patients (odds ratio, OR = 33.111, 95 % confidence interval, CI = 11.912-92.040). The expression of HIF-1α correlated with the depth of invasion (OR = 1.701, 95 % CI = 1.076-4.705), clinical TNM stage (OR = 2.160, 95%CI = 1.516-3.077), as well as lymph node metastasis (OR = 2.393, 95 % CI = 1.319-4.344), regardless of differentiation grading (OR = 1.185, 95 % CI = 0.859-1.635). Furthermore, there was a significant association of increased HIF-1α expression with poorer radiochemotherapy outcomes, 2-year overall survival (OR = 0.219, 95 % CI = 0.104-0.461) and survival (OR = 0.320, 95 % CI = 0.115-0.887, P < 0.05) in patients with ESCC. In addition, HIF-1α expression correlated with VEGF expression in the ESCCs (OR = 4.635, 95%CI = 2.591-8.292). Increased expression of HIF-1α plays an important role in the malignant biology of ESCC resulting in significantly poorer radiochemotherapy outcomes and 2-year overall survival. HIF-1α expression may be a prognostic factor, as well as a potential target for therapy in patients with ESCC.
Collapse
Affiliation(s)
- Guogui Sun
- Department of Chemoradiology, People's Hospital Affiliated to Hebei United University, Tangshan, 063000,, Hebei Province, China
| | | | | | | |
Collapse
|
44
|
Hisamatsu T, Mabuchi S, Matsumoto Y, Kawano M, Sasano T, Takahashi R, Sawada K, Ito K, Kurachi H, Schilder RJ, Testa JR, Kimura T. Potential role of mTORC2 as a therapeutic target in clear cell carcinoma of the ovary. Mol Cancer Ther 2013; 12:1367-77. [PMID: 23615631 DOI: 10.1158/1535-7163.mct-12-1185] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The goal of this study was to examine the role of mTOR complex 2 (mTORC2) as a therapeutic target in ovarian clear cell carcinoma (CCC), which is regarded as an aggressive, chemoresistant histologic subtype. Using tissue microarrays of 98 primary ovarian cancers [52 CCCs and 46 serous adenocarcinomas (SAC)], activation of mTORC2 was assessed by immunohistochemistry. Then, the growth-inhibitory effect of mTORC2-targeting therapy, as well as the role of mTORC2 signaling as a mechanism for acquired resistance to the mTOR complex 1 (mTORC1) inhibitor RAD001 in ovarian CCC, were examined using two pairs of RAD001-sensitive parental (RMG2 and HAC2) and RAD001-resistant CCC cell lines (RMG2-RR and HAC2-RR). mTORC2 was more frequently activated in CCCs than in SACs (71.2% vs. 45.7%). Simultaneous inhibition of mTORC1 and mTORC2 by AZD8055 markedly inhibited the proliferation of both RAD001-sensitive and -resistant cells in vitro. Treatment with RAD001 induced mTORC2-mediated AKT activation in RAD001-sensitive CCC cells. Moreover, increased activation of mTORC2-AKT signaling was observed in RAD001-resistant CCC cells compared with the respective parental cells. Inhibition of mTORC2 during RAD001 treatment enhanced the antitumor effect of RAD001 and prevented CCC cells from acquiring resistance to RAD001. In conclusion, mTORC2 is frequently activated, and can be a promising therapeutic target, in ovarian CCCs. Moreover, mTORC2-targeted therapy may be efficacious in a first-line setting as well as for second-line treatment of recurrent disease developing after RAD001-treatment.
Collapse
Affiliation(s)
- Takeshi Hisamatsu
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Fujisue Y, Nakagawa T, Takahara K, Inamoto T, Kiyama S, Azuma H, Asahi M. Induction of erythropoietin increases the cell proliferation rate in a hypoxia-inducible factor-1-dependent and -independent manner in renal cell carcinoma cell lines. Oncol Lett 2013; 5:1765-1770. [PMID: 23833638 PMCID: PMC3701060 DOI: 10.3892/ol.2013.1283] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 02/25/2013] [Indexed: 11/09/2022] Open
Abstract
Erythropoietin (Epo) is a potent inducer of erythropoiesis that is mainly produced in the kidney. Epo is expressed not only in the normal kidney, but also in renal cell carcinomas (RCCs). The aim of the present study was to gain insights into the roles of Epo and its receptor (EpoR) in RCC cells. The study used two RCC cell lines, Caki-1 and SKRC44, in which Epo and EpoR are known to be highly expressed. The proliferation rate and expression level of hypoxia-inducible factor-1α (HIF-1α) were measured prior to and following Epo treatment and under normoxic and hypoxic conditions. To examine whether HIF-1α or Epo were involved in cellular proliferation during hypoxia, these proteins were knocked down using small interfering RNA (siRNA) in Caki-1 and SKRC44 cells. The results demonstrated that Epo enhanced the proliferation of the Caki-1 and SKRC44 cells. HIF-1α expression was increased upon the induction of hypoxia in the Caki-1 cells, but remained unaffected in the SKRC44 cells. The proliferation rate was increased under hypoxic conditions in the Caki-1 cells, but was decreased in the SKRC44 cells. Under hypoxic conditions, the proliferation of the Caki-1 cells was significantly reduced by the knock-down of HIF-1α or Epo, while the proliferation of the SKRC44 cells was significantly suppressed by the knock-down of Epo, but not HIF-1α. In conclusion, these data suggest that the induction of Epo may accelerate the proliferation of the RCC cell lines in either a HIF-1α-dependent or -independent manner.
Collapse
Affiliation(s)
- Yutaka Fujisue
- Departments of Urology, Faculty of Medicine, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | | | | | | | | | | | | |
Collapse
|
46
|
Kim K, Park WY, Kim JY, Sol MY, Shin DH, Park DY, Lee CH, Lee JH, Choi KU. Prognostic Relevance of the Expression of CA IX, GLUT-1, and VEGF in Ovarian Epithelial Cancers. KOREAN JOURNAL OF PATHOLOGY 2012; 46:532-40. [PMID: 23323103 PMCID: PMC3540330 DOI: 10.4132/koreanjpathol.2012.46.6.532] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 09/12/2012] [Accepted: 09/17/2012] [Indexed: 12/27/2022]
Abstract
Background Tumor hypoxia is associated with malignant progression and treatment resistance. Hypoxia-related factors, such as carbonic anhydrase IX (CA IX), glucose transporter-1 (GLUT-1), and vascular endothelial growth factor (VEGF) permit tumor cell adaptation to hypoxia. We attempted to elucidate the correlation of these markers with variable clinicopathological factors and overall prognosis. Methods Immunohistochemistry for CA IX, GLUT-1, and VEGF was performed on formalin-fixed, paraffin-embedded tissues from 125 cases of ovarian epithelial cancer (OEC). Results CA IX expression was significantly associated with an endometrioid and mucinous histology, nuclear grade, tumor necrosis, and mitosis. GLUT-1 expression was associated with tumor necrosis and mitosis. VEGF expression was correlated only with disease recurrence. Expression of each marker was not significant in terms of overall survival in OECs; however, there was a significant correlation between poor overall survival rate and high coexpression of these markers. Conclusions The present study suggests that it is questionable whether CA IX, GLUT-1, or VEGF can be used alone as independent prognostic factors in OECs. Using at least two markers helps to predict patient outcomes in total OECs. Moreover, the inhibition of two target gene combinations might prove to be a novel anticancer therapy.
Collapse
Affiliation(s)
- Kyungbin Kim
- Department of Pathology, Pusan National University Hospital, Pusan National University School of Medicine, Yangsan, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Kato M, Yamamoto S, Takano M, Matsubara O, Furuya K. Aberrant expression of the mammalian target of rapamycin, hypoxia-inducible factor-1α, and glucose transporter 1 in the development of ovarian clear-cell adenocarcinoma. Int J Gynecol Pathol 2012; 31:254-63. [PMID: 22498943 DOI: 10.1097/pgp.0b013e318237d66c] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Ovarian clear-cell adenocarcinoma (CCA) is known to be a type of cancer in humans with a high frequency of expression of the mammalian target of rapamycin (mTOR), hypoxia-inducible factor-1 (HIF-1), and glucose transporter 1 (Glut1). In this study, we aimed to determine how these alterations contribute to tumor development of CCAs. Immunohistochemical expressions of phosphorylated-mTOR (p-mTOR), HIF-1α, and Glut1 were analyzed in 36 CCAs and 60 coexistent putative precursor lesions: 19 nonatypical and 16 atypical endometriotic lesions, and 11 benign and 14 borderline clear-cell adenofibroma (CCAF) components. Twenty-one cases with solitary endometriosis were also examined. The frequencies of immunopositivity for p-mTOR (in cytoplasm or nucleus), HIF-1α (in nucleus), and Glut1 increased in accordance with higher cytological atypia in the putative precursors: 58%, 5%, and 16% in the nonatypical endometriosis; 63%, 37%, and 50% in the atypical endometriosis; 77%, 95%, and 95% in the endometriosis-associated CCAs; 27%, 0%, and 0% in the benign-CCAF components; 64%, 79%, and 43% in the borderline CCAF components; and 71%, 100%, and 93% in the CCAF-associated CCAs, respectively. p-mTOR, HIF-1α (in the nucleus), and Glut1 were positive in 10%, 5%, and 19% of the solitary endometriosis, respectively. In the putative precursor lesions coexisting with CCA, a strong correlation in the expression between p-mTOR and HIF-1α and between HIF-1α and Glut1 was identified. Expressions of p-mTOR, HIF-1α, and Glut1 have already been evident in the putative precursor lesions of CCA, and these alterations cumulatively occur in the development of ovarian CCA.
Collapse
Affiliation(s)
- Masafumi Kato
- Departments of Obstetrics and Gynecology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | | | | | | | | |
Collapse
|
48
|
van der Bilt AR, van Scheltinga AGT, Timmer-Bosscha H, Schröder CP, Pot L, Kosterink JG, van der Zee AG, Lub-de Hooge MN, de Jong S, de Vries EG, Reyners AK. Measurement of Tumor VEGF-A Levels with 89Zr-Bevacizumab PET as an Early Biomarker for the Antiangiogenic Effect of Everolimus Treatment in an Ovarian Cancer Xenograft Model. Clin Cancer Res 2012; 18:6306-14. [DOI: 10.1158/1078-0432.ccr-12-0406] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
49
|
Takano M, Tsuda H, Sugiyama T. Clear cell carcinoma of the ovary: is there a role of histology-specific treatment? JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2012; 31:53. [PMID: 22655678 PMCID: PMC3405444 DOI: 10.1186/1756-9966-31-53] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 06/01/2012] [Indexed: 11/10/2022]
Abstract
Several clinical trials to establish standard treatment modality for ovarian cancers included a high abundance of patients with serous histologic tumors, which were quite sensitive to platinum-based chemotherapy. On the other hand, ovarian tumor with rare histologic subtypes such as clear cell or mucinous tumors have been recognized to show chemo-resistant phenotype, leading to poorer prognosis. Especially, clear cell carcinoma of the ovary (CCC) is a distinctive tumor, deriving from endometriosis or clear cell adenofibroma, and response rate to platinum-based therapy is extremely low. It was implied that complete surgical staging enabled us to distinguish a high risk group of recurrence in CCC patients whose disease was confined to the ovary (pT1M0); however, complete surgical staging procedures could not lead to improved survival. Moreover, the status of peritoneal cytology was recognized as an independent prognostic factor in early-staged CCC patients, even after complete surgical staging. In advanced cases with CCC, the patients with no residual tumor had significantly better survival than those with the tumor less than 1 cm or those with tumor diameter more than 1 cm. Therefore, the importance of achieving no macroscopic residual disease at primary surgery is so important compared with other histologic subtypes. On the other hand, many studies have shown that conventional platinum-based chemotherapy regimens yielded a poorer prognosis in patients with CCC than in patients with serous subtypes. The response rate by paclitaxel plus carboplatin (TC) was slightly higher, ranging from 22% to 56%, which was not satisfactory enough. Another regimen for CCC tumors is now being explored: irinotecan plus cisplatin, and molecular targeting agents. In this review article, we discuss the surgical issues for early-staged and advanced CCC including possibility of fertility-sparing surgery, and the chemotherapy for CCC disease.
Collapse
Affiliation(s)
- Masashi Takano
- Department of Obstetrics and Gynecology, National Defense Medical College, Tokorozawa, Saitama, Japan.
| | | | | |
Collapse
|
50
|
del Carmen MG, Birrer M, Schorge JO. Clear cell carcinoma of the ovary: a review of the literature. Gynecol Oncol 2012; 126:481-90. [PMID: 22525820 DOI: 10.1016/j.ygyno.2012.04.021] [Citation(s) in RCA: 185] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 04/12/2012] [Accepted: 04/16/2012] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Different histologic types of epithelial ovarian cancer may represent different diseases with unique clinical and molecular characteristics. Clear cell carcinoma (CCC) of the ovary has been reported as having a worse prognosis than high grade serous epithelial ovarian cancer (EOC). This article critically reviews the literature pertinent to the pathology, pathogenesis, diagnosis, management, and outcome of patients with ovarian CCC. METHODS MEDLINE was searched for all research articles published in English between January 01, 1977 and January 30, 2012 which reported on patients diagnosed with ovarian CCC. Given the rarity of this tumor, studies were not limited by design or number of reported patients. RESULTS Ovarian CCC tumors represent 5-25% of ovarian cancers. Its histologic diagnosis can be challenging, resulting often times in misclassification of these tumors. Ovarian CCC tends to present at earlier stages and has been associated with endometriosis, ARID1A and PIK3CA mutations. When compared to stage-matched controls, patients with early-stage ovarian CCCs may have a better prognosis than patients with high-grade serous tumors. For those with advanced stage disease, high-grade serous histology confers a better prognosis than ovarian CCC. Patients with Stage IC-IV have a relatively poor prognosis and efforts should center in discovery of more effective treatment strategies. CONCLUSIONS Ovarian CCC is a biologically distinct entity, different from high-grade serous EOC. Future studies should explore the role of targeted therapies in the management of ovarian CCC.
Collapse
Affiliation(s)
- Marcela G del Carmen
- Division of Gynecologic Oncology, Vincent Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | | | | |
Collapse
|