1
|
Putnová I, Putnová BM, Hurník P, Štembírek J, Buchtová M, Kolísková P. Primary cilia-associated signalling in squamous cell carcinoma of head and neck region. Front Oncol 2024; 14:1413255. [PMID: 39234399 PMCID: PMC11372790 DOI: 10.3389/fonc.2024.1413255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 07/29/2024] [Indexed: 09/06/2024] Open
Abstract
Squamous cell carcinoma (SCC) of the head and neck originates from the mucosal lining of the upper aerodigestive tract, including the lip, tongue, nasopharynx, oropharynx, larynx and hypopharynx. In this review, we summarise what is currently known about the potential function of primary cilia in the pathogenesis of this disease. As primary cilia represent a key cellular structure for signal transduction and are related to cell proliferation, an understanding of their role in carcinogenesis is necessary for the design of new treatment approaches. Here, we introduce cilia-related signalling in head and neck squamous cell carcinoma (HNSCC) and its possible association with HNSCC tumorigenesis. From this point of view, PDGF, EGF, Wnt and Hh signalling are discussed as all these pathways were found to be dysregulated in HNSCC. Moreover, we review the clinical potential of small molecules affecting primary cilia signalling to target squamous cell carcinoma of the head and neck area.
Collapse
Affiliation(s)
- Iveta Putnová
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Department of Anatomy, Histology and Embryology, University of Veterinary Sciences Brno, Brno, Czechia
| | - Barbora Moldovan Putnová
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Department of Pathological Morphology and Parasitology, University of Veterinary Sciences Brno, Brno, Czechia
| | - Pavel Hurník
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Institute of Molecular and Clinical Pathology and Medical Genetics, University Hospital Ostrava, Ostrava, Czechia
- Institute of Molecular and Clinical Pathology and Medical Genetics, Faculty of Medicine, University of Ostrava, Ostrava, Czechia
| | - Jan Štembírek
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Department of Maxillofacial Surgery, University Hospital Ostrava, Ostrava, Czechia
| | - Marcela Buchtová
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Petra Kolísková
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| |
Collapse
|
2
|
Pandey P, Khan F, Upadhyay TK, Seungjoon M, Park MN, Kim B. New insights about the PDGF/PDGFR signaling pathway as a promising target to develop cancer therapeutic strategies. Biomed Pharmacother 2023; 161:114491. [PMID: 37002577 DOI: 10.1016/j.biopha.2023.114491] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 02/20/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023] Open
Abstract
Numerous cancers express platelet-derived growth factors (PDGFs) and PDGF receptors (PDGFRs). By directly stimulating tumour cells in an autocrine manner or by stimulating tumour stromal cells in a paracrine manner, the platelet-derived growth factor (PDGF)/platelet-derived growth factor receptor (PDGFR) pathway is crucial in the growth and spread of several cancers. To combat hypoxia in the tumour microenvironment, it encourages angiogenesis. A growing body of experimental data shows that PDGFs target malignant cells, vascular cells, and stromal cells to modulate tumour growth, metastasis, and the tumour microenvironment. To combat medication resistance and enhance patient outcomes in cancers, targeting the PDGF/PDGFR pathway is a viable therapeutic approach. There have been reports of anomalies in the PDGF pathway, including the gain of function point mutations, activating chromosomal translocations, or overexpression or amplification of PDGF receptors (PDGFRs). As a result, it has been shown that targeting the PDGF/PDGFR signaling pathway is an effective method for treating cancer. As a result, this study will concentrate on the regulation of the PDGF/PDGFR signaling system, in particular the current methods and inhibitors used in cancer treatment, as well as the associated therapeutic advantages and side effects.
Collapse
Affiliation(s)
- Pratibha Pandey
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida, UP, India
| | - Fahad Khan
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida, UP, India.
| | - Tarun Kumar Upadhyay
- Department of Biotechnology, Parul Institute of Applied Sciences and Centre of Research for Development, Parul University, Vadodara 391760, India
| | - Moon Seungjoon
- Chansol Hospital of Korean Medicine, 290, Buheung-ro, Bupyeong-gu, Incheon 21390, Republic of Korea; Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea; Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea; Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
3
|
Li S, Yang H, Li W, Liu JY, Ren LW, Yang YH, Ge BB, Zhang YZ, Fu WQ, Zheng XJ, Du GH, Wang JH. ADH1C inhibits progression of colorectal cancer through the ADH1C/PHGDH /PSAT1/serine metabolic pathway. Acta Pharmacol Sin 2022; 43:2709-2722. [PMID: 35354963 PMCID: PMC9525271 DOI: 10.1038/s41401-022-00894-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 02/27/2022] [Indexed: 12/22/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer in men and the second most common cancer in women worldwide. CRC is the second leading cause of cancer-related deaths. Although some progress in the treatment of CRC has been achieved, the molecular mechanism of CRC is still unclear. In this study, alcohol dehydrogenase 1C(ADH1C) was first identified as a target gene closely associated with the development of CRC by the comprehensive application of transcriptomics, proteomics, metabonomics and in silico analysis. The ADH1C mRNA and protein expression in CRC cell lines and tumor tissues was lower than that in normal intestinal epithelial cell lines and healthy tissues. Overexpression of ADH1C inhibited the growth, migration, invasion and colony formation of CRC cell lines and prevented the growth of xenograft tumors in nude mice. The inhibitory effects of ADH1C on CRC cells in vitro were exerted by reducing the expression of PHGDH/PSAT1 and the serine level. This inhibition could be partially reversed by adding serine to the culture medium. These results showed that ADH1C is a potential drug target in CRC.
Collapse
Affiliation(s)
- Sha Li
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Hong Yang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Wan Li
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Jin-Yi Liu
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Li-Wen Ren
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Yi-Hui Yang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Bin-Bin Ge
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Yi-Zhi Zhang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Wei-Qi Fu
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Xiang-Jin Zheng
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Guan-Hua Du
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China.
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China.
| | - Jin-Hua Wang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China.
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
4
|
Wan X, Zhou M, Huang F, Zhao N, Chen X, Wu Y, Zhu W, Ni Z, Jin F, Wang Y, Hu Z, Chen X, Ren M, Zhang H, Zha X. AKT1-CREB stimulation of PDGFRα expression is pivotal for PTEN deficient tumor development. Cell Death Dis 2021; 12:172. [PMID: 33568640 PMCID: PMC7876135 DOI: 10.1038/s41419-021-03433-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 12/12/2022]
Abstract
As evidenced by the behavior of loss-of-function mutants of PTEN in the context of a gain-of-function mutation of AKT1, the PTEN-AKT1 signaling pathway plays a critical role in human cancers. In this study, we demonstrated that a deficiency in PTEN or activation of AKT1 potentiated the expression of platelet-derived growth factor receptor α (PDGFRα) based on studies on Pten-/- mouse embryonic fibroblasts, human cancer cell lines, the hepatic tissues of Pten conditional knockout mice, and human cancer tissues. Loss of PTEN enhanced PDGFRα expression via activation of the AKT1-CREB signaling cascade. CREB transactivated PDGFRα expression by direct binding of the promoter of the PDGFRα gene. Depletion of PDGFRα attenuated the tumorigenicity of Pten-null cells in nude mice. Moreover, the PI3K-AKT signaling pathway has been shown to positively correlate with PDGFRα expression in multiple cancers. Augmented PDGFRα was associated with poor survival of cancer patients. Lastly, combination treatment with the AKT inhibitor MK-2206 and the PDGFR inhibitor CP-673451 displayed synergistic anti-tumor effects. Therefore, activation of the AKT1-CREB-PDGFRα signaling pathway contributes to the tumor growth induced by PTEN deficiency and should be targeted for cancer treatment.
Collapse
Affiliation(s)
- Xiaofeng Wan
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
- Department of Laboratory, Cancer Hospital, Chinese Academy of Sciences, Hefei, China
| | - Meng Zhou
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Fuqiang Huang
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Na Zhao
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xu Chen
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Yuncui Wu
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Wanhui Zhu
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhaofei Ni
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Fuquan Jin
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Yani Wang
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Zhongdong Hu
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xianguo Chen
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Min Ren
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hongbing Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Xiaojun Zha
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China.
| |
Collapse
|
5
|
Chan KKL, Ngu SF, Chu MMY, Tse KY, Ngan HYS. Tamoxifen use in recurrent ovarian cancer in a Chinese population: A 15 -year clinical experience in a tertiary referral center. Asia Pac J Clin Oncol 2020; 17:338-342. [PMID: 33079469 PMCID: PMC8359459 DOI: 10.1111/ajco.13478] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 09/05/2020] [Indexed: 12/30/2022]
Abstract
Aim To review the clinical use and the effectiveness of tamoxifen in patients with advanced or recurrent ovarian cancer. Methods A retrospective review of clinical records was conducted in patients who received tamoxifen for the treatment of ovarian cancer between 2002 and 2016. We reviewed the clinical setting that it was given, duration of use, patients' tolerability, clinical benefit and progression‐free survival. We also attempted to identify predictive markers for response. Results A total of 92 patients received tamoxifen during this 15‐year period. The patients received a median of 2.5 lines of chemotherapy before switching to tamoxifen, and they remained on tamoxifen for a median of 5.6 months (range 0–85 months), with 24 patients receiving it for more than 12 months. Seventy‐six patients continued on tamoxifen for more than 2 months. In this group, 75 patients had an evaluable response, either by CA 125 or clinically and clinical benefit rate (defined as complete, partial response and static disease) was seen in 42 patients (56%), with majority of patients having static disease. The median progression‐free survival was 5.3 months (95% confidence interval, 2.6–8.1). Tamoxifen was well tolerated. Hormone receptor status was not demonstrated to predict response. Conclusion Patients with advanced ovarian cancer who have failed previous lines of chemotherapy may achieve static disease with tamoxifen with minimal side effects. Tamoxifen may still have a role in the era of molecular target therapy.
Collapse
Affiliation(s)
- Karen Kar Loen Chan
- Department of Obstetrics and Gynaecology, University of Hong Kong, Professorial Block, Queen Mary Hospital, Hong Kong
| | - Siew Fei Ngu
- Department of Obstetrics and Gynaecology, University of Hong Kong, Professorial Block, Queen Mary Hospital, Hong Kong
| | - Mandy Man Yee Chu
- Department of Obstetrics and Gynaecology, University of Hong Kong, Professorial Block, Queen Mary Hospital, Hong Kong
| | - Ka Yu Tse
- Department of Obstetrics and Gynaecology, University of Hong Kong, Professorial Block, Queen Mary Hospital, Hong Kong
| | - Hextan Yuen Sheung Ngan
- Department of Obstetrics and Gynaecology, University of Hong Kong, Professorial Block, Queen Mary Hospital, Hong Kong
| |
Collapse
|
6
|
Zoledronic Acid Abrogates Restraint Stress-Induced Macrophage Infiltration, PDGF-AA Expression, and Ovarian Cancer Growth. Cancers (Basel) 2020; 12:cancers12092671. [PMID: 32962103 PMCID: PMC7563308 DOI: 10.3390/cancers12092671] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 09/16/2020] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Biobehavioral disorders can negatively impact patients with ovarian cancer. Growing evidence suggests that chronic stress can promote tumor progression, the release of inflammatory mediators, and macrophage infiltration into the tumor. However, the role of stress hormones in regulating cancer cell/macrophage crosstalk remains unclear. This study aimed to assess the role of stress hormone-stimulated macrophages in modulating inflammatory networks and ovarian cancer biology. Our data show that stress hormones induced secretion of inflammatory proteins in ovarian cancer cell/macrophage co-cultures. Furthermore, we show that restraint stress leads to cancer growth, macrophage infiltration, and PDGF-AA protein expression in animal models of ovarian cancer. Conversely, zoledronic acid was able to prevent the effects of restraint stress on ovarian cancer growth. Overall, our data suggest a role for stress hormone-stimulated macrophages in ovarian cancer progression and suggest the involvement of PDGF-AA as a key mediator of this process. Abstract Multiple studies suggest that chronic stress accelerates the growth of existing tumors by activating the sympathetic nervous system. Data suggest that sustained adrenergic signaling can induce tumor growth, secretion of pro-inflammatory cytokines, and macrophage infiltration. Our goal was to study the role of adrenergic-stimulated macrophages in ovarian cancer biology. Cytokine arrays were used to assess the effect of adrenergic stimulation in pro-tumoral cytokine networks. An orthotopic model of ovarian cancer was used to assess the in vivo effect of daily restraint stress on tumor growth and adrenergic-induced macrophages. Cytokine analyses showed that adrenergic stimulation modulated pro-inflammatory cytokine secretion in a SKOV3ip1 ovarian cancer cell/U937 macrophage co-culture system. Among these, platelet-derived growth factor AA (PDGF-AA), epithelial cell-derived neutrophil-activating peptide (ENA-78), Angiogenin, vascular endothelial growth factor (VEGF), granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-5 (IL-5), Lipocalin-2, macrophage migration inhibitory factor (MIF), and transferrin receptor (TfR) were upregulated. Enriched biological processes included cytokine-mediated signaling pathways and positive regulation of cell proliferation. In addition, daily restraint stress increased ovarian cancer growth, infiltration of CD68+ macrophages, and expression of PDGF-AA in orthotopic models of ovarian cancer (SKOV3ip1 and HeyT30), while zoledronic acid, a macrophage-depleting agent, abrogated this effect. Furthermore, in ovarian cancer patients, high PDGFA expression correlated with worse outcomes. Here, it is shown that the adrenergic regulation of macrophages and PDGFA might play a role in ovarian cancer progression.
Collapse
|
7
|
Sun L, Yang M, Zhang X, Li H, Wu L, Zhang Y, Cai S. Anlotinib combined with etoposide for platinum-resistant recurrent ovarian cancer: A case report. Medicine (Baltimore) 2020; 99:e20053. [PMID: 32443311 PMCID: PMC7253621 DOI: 10.1097/md.0000000000020053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION Platinum-resistant ovarian cancer is characterized by its poor prognosis and limited treatment options. Angiogenesis plays a fundamental role in the development of drug-resistance in ovarian cancer. Anlotinib, a novel oral multi-targeted tyrosine kinase inhibitor which targets a board spectrum of angiogenesis-associated growth factor receptors, has shown promising anti-tumor efficacy in clinical trials. Herein, we report a case of ovarian cancer treated with anlotinib plus etoposide after secondary cytoreductive surgery. PATIENT CONCERNS A 45-year-old female with primary platinum-resistant ovarian cancer who progressed rapidly after the first cytoreductive surgery, the second cytoreductive surgery, and several lines of treatment. The patient refused to receive intravenous chemotherapy any more. DIAGNOSIS Primary platinum-resistant ovarian cancer. INTERVENTIONS The oral combination treatment of anlotinib (12 mg, qd) and etoposide (100 mg, qd) were delivered. OUTCOMES Finally, the patient was responsive to the orally treatment of anlotinib combined with etoposide. The patient has been alive with no evidence of disease progression for 18 weeks. CONCLUSION Our case suggests that oral treatment of anlotinib combined with etoposide which is acceptable and convenient, may be an additional option for the management of platinum-resistant ovarian cancer.
Collapse
Affiliation(s)
- Li Sun
- Departments of Gynecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
- Departments of Gynecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital l & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen
| | - Meng Yang
- Departments of Gynecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital l & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen
| | - Xuan Zhang
- Departments of Gynecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital l & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen
| | - Hua Li
- Departments of Gynecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital l & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen
| | - Lingying Wu
- Departments of Gynecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Yuzi Zhang
- The Medical Department, 3D Medicines Inc, Shanghai, People's Republic of China
| | - Shangli Cai
- The Medical Department, 3D Medicines Inc, Shanghai, People's Republic of China
| |
Collapse
|
8
|
Canzler U, Lück HJ, Neuser P, Sehouli J, Burges A, Harter P, Schmalfeldt B, Aminossadati B, Mahner S, Kommoss S, Wimberger P, Pfisterer J, de Gregorio N, Hasenburg A, Gropp-Meier M, El-Balat A, Jackisch C, du Bois A, Meier W, Wagner U. Prognostic role of thrombocytosis in recurrent ovarian cancer: a pooled analysis of the AGO Study Group. Arch Gynecol Obstet 2020; 301:1267-1274. [PMID: 32277253 DOI: 10.1007/s00404-020-05529-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/28/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE Although thrombocytosis in patients with primary ovarian cancer has been widely investigated, there are only very few data about the role of thrombocytosis in recurrent ovarian cancer. The aim of our study was to investigate the impact of pretreatment thrombocytosis prior to chemotherapy on clinical outcome in patients with recurrent platinum eligible ovarian cancer. METHODS In our retrospective analysis we included 300 patients who were treated by AGO Study Group Centers within three prospective, randomized phase-III-trials. All patients included had been treatment-free for at least 6 months after platinum-based chemotherapy. We excluded patients who underwent secondary cytoreductive surgery before randomization to the trial. Thrombocytosis was defined as a platelet count of ≥ 400⋅109/L. RESULTS Pretreatment thrombocytosis was present in 37 out of 300 (12.3%) patients. Patients with thrombocytosis responded statistically significantly less to chemotherapy (overall response rate 35.3% and 41.6%, P = 0.046). The median progression-free survival (PFS) for patients with thrombocytosis was 6.36 months compared to 9.00 months for patients without thrombocytosis (hazard ratio [HR] = 1.19, 95% confidence interval [CI] = 0.84-1.69, P = 0.336). Median overall survival (OS) of patients with thrombocytosis was 16.33 months compared to 23.92 months of patients with a normal platelet count (HR = 1.46, 95% CI = 1.00-2.14, P = 0.047). CONCLUSIONS The present analysis suggests that pretreatment thrombocytosis is associated with unfavorable outcome with regard to response to chemotherapy and overall survival in recurrent ovarian cancer.
Collapse
Affiliation(s)
- Ulrich Canzler
- Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany. .,National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.
| | | | - Petra Neuser
- Coordinating Center for Clinical Trials, Philipps University Marburg, Marburg, Germany
| | - Jalid Sehouli
- Department for Gynecology and Center for Oncological Surgery, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Alexander Burges
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Philipp Harter
- Department of Gynecology and Gynecologic Oncology, Kliniken Essen-Mitte, Evangelische Huyssens-Stiftung, Essen, Germany
| | - Barbara Schmalfeldt
- Department of Gynecology and Obstetrics, Klinikum rechts der Isar München, Munich, Germany.,Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Behnaz Aminossadati
- Coordinating Center for Clinical Trials, Philipps University Marburg, Marburg, Germany
| | - Sven Mahner
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany.,Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Kommoss
- Department of Obstetrics and Gynecology, University of Tübingen, Tübingen, Germany
| | - Pauline Wimberger
- Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.,Department of Gynecology and Obstetrics, University Hospital Essen, Essen, Germany
| | - Jacobus Pfisterer
- Department of Gynecology and Obstetrics, Universitätsklinikum Schleswig-Holstein, Kiel, Germany.,Gynecologic Oncology Center, Kiel, Germany
| | | | - Annette Hasenburg
- Department of Gynecology and Gynecologic Oncology, University Hospital Freiburg, Freiburg im Breisgau, Germany.,Department of Obstetrics and Gynecology, University Medical Center, Mainz, Germany
| | - Martina Gropp-Meier
- Department of Gynecology and Obstetrics, Oberschwabenklinik, Krankenhaus St. Elisabeth, Ravensburg, Germany
| | - Ahmed El-Balat
- Department of Gynecology, University of Frankfurt, Frankfurt, Germany
| | - Christian Jackisch
- Department of Obstetrics and Gynecology, Sana Klinikum Offenbach GmbH, Offenbach, Germany
| | - Andreas du Bois
- Department of Gynecology and Gynecologic Oncology, Kliniken Essen-Mitte, Evangelische Huyssens-Stiftung, Essen, Germany
| | - Werner Meier
- Department of Obstetrics and Gynecology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Uwe Wagner
- Department of Gynecology and Obstetrics, University Hospital Giessen and Marburg, Marburg, Germany
| |
Collapse
|
9
|
Miyake K, Kiyuna T, Miyake M, Kawaguchi K, Zhang Z, Wangsiricharoen S, Razmjooei S, Oshiro H, Higuchi T, Li Y, Nelson SD, Murakami T, Hiroshima Y, Kumamoto T, Matsuyama R, Bouvet M, Singh SR, Chawla SP, Endo I, Hoffman RM. Gemcitabine combined with docetaxel precisely regressed a recurrent leiomyosarcoma peritoneal metastasis in a patient-derived orthotopic xenograft (PDOX) model. Biochem Biophys Res Commun 2019; 509:1041-1046. [PMID: 30660363 DOI: 10.1016/j.bbrc.2019.01.046] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 01/08/2019] [Indexed: 12/18/2022]
Abstract
There are no effective treatments for leiomyosarcoma (LMS) spreading intraabdominally. The aim of this study was to develop precision chemotherapy for recurrent peritoneal LMS metastases in a patient-derived orthotopic xenograft (PDOX) model. The LMS PDOX models were established orthotopically on the dome of the bladder of nude mice. The LMS PDOX models were randomized into 6 groups when the tumor volume reached 80 mm3: G1: untreated control; G2: doxorubicin (DOX) (DOX: i.p., 3 mg/kg, weekly, 3 weeks); G3: DOX combined with olaratumab (OLA) (DOX: i.p., 3 mg/kg, weekly, 3 weeks; OLA: i.p., 40 mg/kg, 3 times/week, 3 weeks); G4: gemcitabine (GEM) combined with docetaxel (DOC) (GEM: i.p., 100 mg/kg, weekly, 3 weeks; DOC: i.p., 20 mg/kg, weekly, 3 weeks); G5: pazopanib (PAZ) (PAZ: p.o., 100 mg/kg, daily, 3 weeks); G6: palbociclib (PAL) (PAL: p.o., 100 mg/kg, daily, 3 weeks). All mice were sacrificed on day 22. Body weight was assessed twice a week. Tumor volume was measured on day 0 and day 22. Although all regimens had a significant efficacy compared to the untreated group (P < 0.001), only GEM combined with DOC regressed the tumor significantly (P < 0.001), suggesting GEM combined with DOC has clinical potential for this LMS patient.
Collapse
Affiliation(s)
- Kentaro Miyake
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA; Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tasuku Kiyuna
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | - Masuyo Miyake
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA; Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kei Kawaguchi
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | - Zhiying Zhang
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | | | - Sahar Razmjooei
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | - Hiromichi Oshiro
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | - Takashi Higuchi
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | - Yunfeng Li
- Dept. of Pathology, University of California, Los Angeles, CA, USA
| | - Scott D Nelson
- Dept. of Pathology, University of California, Los Angeles, CA, USA
| | - Takashi Murakami
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yukihiko Hiroshima
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takafumi Kumamoto
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Ryusei Matsuyama
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Michael Bouvet
- Department of Surgery, University of California, San Diego, CA, USA
| | - Shree Ram Singh
- Basic Research Laboratory, National Cancer Institute, Frederick, MD, USA.
| | - Sant P Chawla
- Sarcoma Oncology Center, 2811 Wilshire Blvd, Suite 414, Santa Monica, CA, 90403, USA
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Robert M Hoffman
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA.
| |
Collapse
|
10
|
McGuire WP, Penson RT, Gore M, Herraez AC, Peterson P, Shahir A, Ilaria R. Randomized phase II study of the PDGFRα antibody olaratumab plus liposomal doxorubicin versus liposomal doxorubicin alone in patients with platinum-refractory or platinum-resistant advanced ovarian cancer. BMC Cancer 2018; 18:1292. [PMID: 30591028 PMCID: PMC6307114 DOI: 10.1186/s12885-018-5198-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 12/09/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Olaratumab is a platelet-derived growth factor receptor-α (PDGFRα)-targeting monoclonal antibody blocking PDGFRα signaling. PDGFRα expression is associated with a more aggressive phenotype and poor ovarian cancer outcomes. This randomized, open label phase II study evaluated olaratumab plus liposomal doxorubicin compared with liposomal doxorubicin alone in advanced ovarian cancer patients. METHODS Patients with platinum-refractory or platinum-resistant advanced ovarian cancer were randomized 1:1 to receive liposomal doxorubicin (40 mg/m2, intravenous infusion) administered every 4 weeks with or without olaratumab (20 mg/kg, IV infusion) every 2 weeks. Patients were stratified based on prior response to platinum therapy (refractory vs resistant). The primary efficacy endpoint was progression-free survival (PFS). Secondary endpoints included overall survival (OS), objective response rate, duration of response, and safety. RESULTS A total of 123 patients were treated (62 olaratumab+liposomal doxorubicin; 61 liposomal doxorubicin). Median PFS was 4.2 months for olaratumab+liposomal doxorubicin and 4.0 months for liposomal doxorubicin (stratified hazard ratio [HR] = 1.043; 95% confidence interval [CI] 0.698-1.558; p = 0.837). Median OS was 16.6 months and 16.2 months in the olaratumab+liposomal doxorubicin and liposomal doxorubicin arms, respectively (HR = 1.098; 95% CI 0.71-1.71). In the platinum-refractory subgroup, median PFS was 5.5 months (95% CI 1.6-9.2) and 3.7 months (95% CI 1.9-9.2) in the olaratumab+liposomal doxorubicin (n = 15) and liposomal doxorubicin arms (n = 16), respectively (HR = 0.85; 95% CI 0.38-1.91). Overall, 59.7% (olaratumab+liposomal doxorubicin) and 65.6% (liposomal doxorubicin) of patients reported grade ≥ 3 adverse events regardless of causality. The most common treatment-emergent adverse events (all grades) regardless of causality were fatigue related (61%), nausea (57%), and constipation (52%) with olaratumab+liposomal doxorubicin and nausea (64%), fatigue related (62%), and mucositis (46%) with liposomal doxorubicin. CONCLUSIONS The addition of olaratumab to liposomal doxorubicin did not result in significant prolongation of PFS or OS in platinum-resistant or platinum-refractory ovarian cancer. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT00913835 ; registered June 2, 2009.
Collapse
Affiliation(s)
- William P McGuire
- Virginia Commonwealth University, 1201 E Marshall St, Room 11-210, Richmond, VA, 23298, USA.
| | - Richard T Penson
- Massachusetts General Hospital, Yawkey 9-064, 32 Fruit St, Boston, MA, 02114, USA
| | - Martin Gore
- The Royal Marsden Hospital, Fulham Road, London, SW3 6JJ, UK
| | | | | | - Ashwin Shahir
- Eli Lilly and Company, Lilly UK, EMC Building, Erl Wood Manor, Windlesham, Surrey, GU20 6PH, UK
| | - Robert Ilaria
- Eli Lilly and Company, Indianapolis, IN, USA.,, Celgene Corporation, 86 Morris Ave, Summit, NJ, 07901, USA
| |
Collapse
|
11
|
Lu L, Fu X, Li Z, Qiu Y, Li W, Zhou Z, Xue W, Wang Y, Jin M, Zhang M. Platelet-derived growth factor receptor alpha (PDGFRα) is overexpressed in NK/T-cell lymphoma and mediates cell survival. Biochem Biophys Res Commun 2018; 504:525-531. [PMID: 30201265 DOI: 10.1016/j.bbrc.2018.08.181] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 08/28/2018] [Indexed: 11/19/2022]
Abstract
Nasal-type natural killer/T-cell lymphoma (NKTCL) is a subtype of non-Hodgkin lymphoma (NHL) that is clinically aggressive and has a poor prognosis. Platelet-derived growth factor receptors (PDGFRs) and their ligands (PDGFs) play important roles in angiogenesis, cancer cell proliferation, survival, migration and poor prognosis in various tumours. However, the significance of PDGFRs in NKTCL remains unknown. Herein, the present study aimed to investigate the important role of PDGFRα in pathogenesis, progression and prognisis of NKTCL. Firstly, we performed immunohistochemical staining, qRT-PCR and western blotting to determine PDGFRα expression in formalin-fixed, paraffin-embedded tissue sections from 78 NKTCL cases and in cell lines. Secondly, correlations between PDGFRα expression and NKTCL clinical parameters and prognosis were analysed. Moreover, a biological assessment of PDGFRα blockade in two NKTCL cell lines was conducted through proliferation assay, cell-cycle evaluation and apoptosis detection by flow cytometry analyses. Furthermore, we detected in vivo activity of imatinib in mouse model of NKTCL. We found that the expression of PDGFRα was significantly higher in NKTCL tissues compared to the reactive lymphoid hyperplasia of the nasopharynx (P = 0.028). High PDGFRα expression was strongly associated with a high LDH level (P = 0.028) and III-IV stage (P = 0.013). NKTCL patients with high PDGFRα expression displayed a reduced median overall survival time and progression-free survival time when compared with those with low PDGFRα expression (P = 0.011, P = 0.005, respectively). Cox multivariate analysis showed that III-IV stage (P = 0.024) and high PDGFRα expression (P = 0.003) were independent prognostic factors in NKTCL patients. Biological assessment assays in two NKTCL cell lines revealed that a specific PDGFR antagonist, imatinib, inhibited cell viability, blocked cell cycle progression at G0/G1 stage and induced apoptosis. Similarly, the in vivo assay showed that imatinib delayed mouse model tumour growth. In conclusion, NKTCL tumour cells have prominent PDGFRα expression, which can serve as a candidate prognostic marker. PDGFR antagonists have significant biological effect on NKTCL and may be useful therapeutic agents for treatment of NKTCL.
Collapse
Affiliation(s)
- Lisha Lu
- Lymphoma Diagnosis and Treatment Center, Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Xiaorui Fu
- Lymphoma Diagnosis and Treatment Center, Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Zhaoming Li
- Lymphoma Diagnosis and Treatment Center, Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Yajuan Qiu
- Lymphoma Diagnosis and Treatment Center, Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Weiming Li
- Lymphoma Diagnosis and Treatment Center, Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Zhiyuan Zhou
- Lymphoma Diagnosis and Treatment Center, Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Weili Xue
- Lymphoma Diagnosis and Treatment Center, Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Yingjun Wang
- Lymphoma Diagnosis and Treatment Center, Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Mengyuan Jin
- Lymphoma Diagnosis and Treatment Center, Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Mingzhi Zhang
- Lymphoma Diagnosis and Treatment Center, Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China.
| |
Collapse
|
12
|
Bonilla L, Oza A, Lheureux S. Emerging growth factor receptor antagonists for ovarian cancer treatment. Expert Opin Emerg Drugs 2018. [PMID: 29528256 DOI: 10.1080/14728214.2018.1446942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
INTRODUCTION Epithelial Ovarian Cancer (EOC) is the most lethal gynecological malignancy. EOC outcomes remain unsatisfactory despite aggressive surgical approach, disease chemo-sensitivity and recent introduction of agents targeting angiogenesis and tumour genome instability. Advances in EOC research have allowed for a tailored treatment approach and accelerated development of novel treatments strategies from bench to bed side, anticipated to improve patient outcomes. Areas covered: Comprehensive review of growth factor receptor antagonists for EOC treatment currently in different stages of development was performed. English peer-reviewed articles and abstracts were searched in MEDLINE, PubMed, Embase and major conferences. We focused on agents that antagonize growth factors promoting sustained proliferative signaling, angiogenesis and evasion of immune destruction blocking the receptor or its stimulating factors. Expert opinion: Receptor signaling has been well characterized for most cancer generating pathways. Growth receptor antagonists are represented by both high receptor affinity monoclonal antibodies as well as tyrosine kinase inhibitors; both are especially effective when a related predictive biomarker of response is identified. Therefore, along with the promising development of novel receptor antagonists or modulators in EOC treatment, targeting essential growth pathways in the tumour and associated microenvironment, is fundamental for biomarker discovery and towards achieving significant improvements in response.
Collapse
Affiliation(s)
- Luisa Bonilla
- a Princess Margaret Cancer Centre , Toronto , Canada
| | - Amit Oza
- a Princess Margaret Cancer Centre , Toronto , Canada
| | | |
Collapse
|
13
|
Dihydroartemisinin selectively inhibits PDGFRα-positive ovarian cancer growth and metastasis through inducing degradation of PDGFRα protein. Cell Discov 2017; 3:17042. [PMID: 29387451 PMCID: PMC5787695 DOI: 10.1038/celldisc.2017.42] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 09/22/2017] [Indexed: 02/06/2023] Open
Abstract
To develop traditional medicines as modern pharmacotherapies, understanding their molecular mechanisms of action can be very helpful. We have recently reported that Artemisinin and its derivatives, which are clinically used anti-malarial drugs, have significant effects against ovarian cancer, but the direct molecular targets and related combination therapy have been unclear. Herein, we report that dihydroartemisinin, one of the most active derivatives of Artemisinin, directly targets platelet-derived growth factor receptor-alpha (PDGFRα) to inhibit ovarian cancer cell growth and metastasis. Dihydroartemisinin directly binds to the intercellular domain of PDGFRα, reducing its protein stability by accelerating its ubiquitin-mediated degradation, which further inactivates downstream phosphoinositide 3-Kinase and mitogen-activated protein kinase pathways and subsequently represses epithelial–mesenchymal transition, inhibiting cell growth and metastasis of PDGFRα-positive ovarian cancer in vitro and in vivo. A combinational treatment reveals that dihydroartemisinin sensitizes ovarian cancer cells to PDGFR inhibitors. Our clinical study also finds that PDGFRα is overexpressed and positively correlated with high grade and metastasis in human ovarian cancer. Considering that Artemisinin compounds are currently clinically used drugs with favorable safety profiles, the results from this study will potentiate their use in combination with clinically used PDGFRα inhibitors, leading to maximal therapeutic efficacy with minimal adverse effects in PDGFRα-positive cancer patients. These findings also shed high light on future development of novel Artemisinin-based targeted therapy.
Collapse
|
14
|
Ravoori MK, Singh SP, Lee J, Bankson JA, Kundra V. In Vivo Assessment of Ovarian Tumor Response to Tyrosine Kinase Inhibitor Pazopanib by Using Hyperpolarized 13C-Pyruvate MR Spectroscopy and 18F-FDG PET/CT Imaging in a Mouse Model. Radiology 2017; 285:830-838. [PMID: 28707963 DOI: 10.1148/radiol.2017161772] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Purpose To assess in a mouse model whether early or late components of glucose metabolism, exemplified by fluorine 18 (18F) fluorodeoxyglucose (FDG) positron emission tomography (PET) and hyperpolarized carbon 13 (13C)-pyruvate magnetic resonance (MR) spectroscopy, can serve as indicators of response in ovarian cancer to multityrosine kinase inhibitor pazopanib. Materials and Methods In this Animal Care and Use Committee approved study, 17 days after the injection of 2 × 106 human ovarian SKOV3 tumors cells into 14 female nude mice, treatment with vehicle or pazopanib (2.5 mg per mouse peroral every other day) was initiated. Longitudinal T2-weighted MR imaging, dynamic MR spectroscopy of hyperpolarized pyruvate, and 18F-FDG PET/computed tomographic (CT) imaging were performed before treatment, 2 days after treatment, and 2 weeks after treatment. Results Pazopanib inhibited ovarian tumor growth compared with control (0.054 g ± 0.041 vs 0.223 g ± 0.112, respectively; six mice were treated with pazopanib and seven were control mice; P < .05). Significantly higher pyruvate-to-lactate conversion (lactate/pyruvate + lactate ratio) was found 2 days after treatment with pazopanib than before treatment (0.46 ± 0.07 vs 0.31 ± 0.14, respectively; P < .05; six tumors after treatment, seven tumors before treatment). This was not observed with the control group or with 18F-FDG PET/CT imaging. Conclusion The findings suggest that hyperpolarized 13C-pyruvate MR spectroscopy may serve as an early indicator of response to tyrosine kinase (angiogenesis) inhibitors such as pazopanib in ovarian cancer even when 18F-FDG PET/CT does not indicate a response. © RSNA, 2017 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Murali K Ravoori
- From the Departments of Cancer Systems Imaging (M.K.R., S.P.S., V.K.), Imaging Physics (J.L., J.A.B.), and Diagnostic Radiology (V.K.), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030
| | - Sheela P Singh
- From the Departments of Cancer Systems Imaging (M.K.R., S.P.S., V.K.), Imaging Physics (J.L., J.A.B.), and Diagnostic Radiology (V.K.), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030
| | - Jaehyuk Lee
- From the Departments of Cancer Systems Imaging (M.K.R., S.P.S., V.K.), Imaging Physics (J.L., J.A.B.), and Diagnostic Radiology (V.K.), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030
| | - James A Bankson
- From the Departments of Cancer Systems Imaging (M.K.R., S.P.S., V.K.), Imaging Physics (J.L., J.A.B.), and Diagnostic Radiology (V.K.), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030
| | - Vikas Kundra
- From the Departments of Cancer Systems Imaging (M.K.R., S.P.S., V.K.), Imaging Physics (J.L., J.A.B.), and Diagnostic Radiology (V.K.), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030
| |
Collapse
|
15
|
Avril S, Dincer Y, Malinowsky K, Wolff C, Gündisch S, Hapfelmeier A, Boxberg M, Bronger H, Becker KF, Schmalfeldt B. Increased PDGFR-beta and VEGFR-2 protein levels are associated with resistance to platinum-based chemotherapy and adverse outcome of ovarian cancer patients. Oncotarget 2017; 8:97851-97861. [PMID: 29228656 PMCID: PMC5716696 DOI: 10.18632/oncotarget.18415] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 05/05/2017] [Indexed: 12/12/2022] Open
Abstract
Despite frequent initial response rates of epithelial ovarian cancer to platinum-based chemotherapy the majority of patients develop drug resistance. Our aim was to evaluate differential expression of signaling-pathway proteins in platinum-sensitive versus platinum-resistant primary epithelial ovarian cancer specimens to identify predictive biomarkers for treatment response. 192 patients were studied comprising of independent training (n = 89) and validation (n = 103) cohorts. Full-length proteins were extracted from paraffin-embedded samples including multiple regions per tumor to account for intratumoral heterogeneity. Quantitative reverse-phase-protein-arrays were used to analyze protein and phospho-protein levels of 41 signaling molecules including growth-factor receptors, AKT and MAPK signaling pathways as well as angiogenesis and cell-adhesion. Platinum-resistant ovarian cancers (56/192) demonstrated significantly higher intratumoral levels of the angiogenesis-associated growth-factor receptors PDGFR-beta and VEGFR2 compared to platinum-sensitive tumors. In addition, patients with high PDGFR-beta expression had significantly shorter overall and progression-free survival (HR 3.6 and 2.4; p < 0.001). The prognostic value of PDGFR-beta and VEGFR2 was confirmed in publicly available microarray-datasets. High intratumoral levels of the angiogenesis-related growth-factor receptors PDGFR-beta and VEGFR2 might serve as novel predictive biomarkers to identify primary resistance to platinum-based chemotherapy. Those ovarian cancer patients might particularly benefit from additional anti-vascular therapy including anti-VEGF antibody or receptor tyrosine-kinase-inhibitor therapy.
Collapse
Affiliation(s)
- Stefanie Avril
- Institute of Pathology, Technische Universität München, Munich, Germany.,Current address: Department of Pathology, Case Western Reserve University School of Medicine, University Hospitals Cleveland Medical Center and Case Comprehensive Cancer Center, Cleveland, Ohio, United States
| | - Yasemin Dincer
- Institute of Pathology, Technische Universität München, Munich, Germany
| | | | - Claudia Wolff
- Institute of Pathology, Technische Universität München, Munich, Germany
| | - Sibylle Gündisch
- Institute of Pathology, Technische Universität München, Munich, Germany
| | - Alexander Hapfelmeier
- Institute of Medical Statistics and Epidemiology, Technische Universität München, Munich, Germany
| | - Melanie Boxberg
- Institute of Pathology, Technische Universität München, Munich, Germany
| | - Holger Bronger
- Department of Obstetrics and Gynecology, Technische Universität München, Munich, Germany
| | | | - Barbara Schmalfeldt
- Department of Obstetrics and Gynecology, Technische Universität München, Munich, Germany.,Current address: Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
16
|
Ekpe-Adewuyi E, Lopez-Campistrous A, Tang X, Brindley DN, McMullen TPW. Platelet derived growth factor receptor alpha mediates nodal metastases in papillary thyroid cancer by driving the epithelial-mesenchymal transition. Oncotarget 2016; 7:83684-83700. [PMID: 27845909 PMCID: PMC5347797 DOI: 10.18632/oncotarget.13299] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 10/22/2016] [Indexed: 01/15/2023] Open
Abstract
Recently platelet derived growth factor receptor-alpha (PDGFRα) was recognized as a potential target to treat aggressive papillary thyroid cancer given its strong association with lymph node metastases. However, it is unclear how PDGFRα potentiates metastases and if it works through the canonical MAPK pathway traditionally linked to PTC oncogenesis. We explored the phenotypic changes driven by PDGFRα activation in human papillary thyroid cancer (PTC) cells and the downstream signalling cascades through which they are effected. We demonstrate that PDGFRα drives an impressive phenotypic change in PTC cell lines as documented by significant cytoskeletal rearrangement, increased migratory potential, and the formation of invadopodia. Cells lacking PDGFRα formed compact and dense spheroids, whereas cells expressing active PDGFRα exhibited invadopodia in three-dimensional culture. To achieve this, active PDGFRα provoked downstream activation of the MAPK/Erk, PI3K/Akt and STAT3 pathways. We further confirmed the role of PDGFRα as a transformative agent promoting the epithelial to mesenchymal transition of PTC cells, through the augmentation of Snail and Slug expression. Crenolanib, a small molecule inhibitor of PDGFRα, suppressed the levels of Snail and Slug and almost completely reversed all the phenotypic changes. We demonstrate that PDGFRα activation is an essential component that drives aggressiveness in PTC cells, and that the signaling pathways are complex, involving not only the MAPK/Erk but also the PI3K/Akt and STAT3 pathways. This argues for upstream targeting of the PDGFRα given the redundancy of oncogenic pathways in PTC, especially in patients whose tumors over-express this tyrosine kinase receptor.
Collapse
Affiliation(s)
| | | | - Xiaoyun Tang
- Signal Transduction Research Group, Department of Biochemistry, University of Alberta, Edmonton, Canada
| | - David N. Brindley
- Signal Transduction Research Group, Department of Biochemistry, University of Alberta, Edmonton, Canada
| | - Todd P. W. McMullen
- Department of Surgery, University of Alberta, Edmonton, Canada
- Division of Surgical Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Canada
| |
Collapse
|
17
|
Hao J, Zhu C, Cao R, Yang X, Ding X, Man Y, Wu X. [Purple-bluish tongue is associated with platelet counts, and the recurrence of epithelial ovarian cancer]. J TRADIT CHIN MED 2016; 36:321-5. [PMID: 27468546 DOI: 10.1016/s0254-6272(16)30044-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To evaluate the relationship between purple-bluish tongue and platelet counts, and further to examine their associations with the recurrence of epithelial ovarian cancer. METHODS A total of 82 epithelial ovarian cancer patients were enrolled in this study. Cluster analysis was used for grouping patients' P(RGB) (Red-R; Green-G; Blue-B; Average percentage of RGB, P(RGB)) values. Receiver operating characteristic (ROC) curve was performed for detecting the diagnostic standard of purple-bluish tongue. Χ2 test was used to assess the relationship between purple-bluish tongue and platelet counts, and the recurrence of epithelial ovarian cancer. The perioperative (preoperative) platelet level was examined with tongue image and disease recurrence. RESULTS Tongue images were classified into two groups basing on P(RGB) values of images by cluster analysis. The numbers of cases in cluster "1" (normal color tongue) was 16 and cluster "2" (purple-bluish tongue) was 66. Two groups of P(RGB) values, classified by cluster analysis, were significantly correlated with vision-based tongue color recognition (Kappa = 0.852, P < 0.001). ROC curve showed that the ratio of P(B) to P(R) had the highest diagnostic value. The sensitivity and the specificity of the ratio of P(B) to P(R) were 95.3% and 88.9% respectively and the optimal cut-off point was 0.71. Purple-bluish tongue was significantly correlated with increased platelet counts (P < 0.001). Both the increased platelet counts (P = 0.01) and purple-bluish tongue were associated with recurrence of epithelial ovarian cancer (P < 0.001). CONCLUSION The ratio of P(B) to P(R) greater than 0.71 could serve as an indicator for purple-bluish tongue diagnosing used in symptom pattern identification in Traditional Chinese Medicine. Purple-bluish tongue, associated with increased platelet counts, was also closely correlated with the recurrence of epithelial ovarian cancer.
Collapse
|
18
|
Bax HJ, Josephs DH, Pellizzari G, Spicer JF, Montes A, Karagiannis SN. Therapeutic targets and new directions for antibodies developed for ovarian cancer. MAbs 2016; 8:1437-1455. [PMID: 27494775 PMCID: PMC5098446 DOI: 10.1080/19420862.2016.1219005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Antibody therapeutics against different target antigens are widely used in the treatment of different malignancies including ovarian carcinomas, but this disease still requires more effective agents. Improved understanding of the biological features, signaling pathways, and immunological escape mechanisms involved in ovarian cancer has emerged in the past few years. These advances, including an appreciation of the cross-talk between cancer cells and the patient's immune system, have led to the identification of new targets. In turn, potential antibody treatments with various mechanisms of action, including immune activation or toxin-delivery, that are directed at these targets have been developed. Here, we identify established as well as novel targets for antibodies in ovarian cancer, and discuss how they may provide fresh opportunities to identify interventions with enhanced therapeutic potential.
Collapse
Affiliation(s)
- Heather J Bax
- a St. John's Institute of Dermatology, Division of Genetics and Molecular Medicine, Faculty of Life Sciences and Medicine, King's College London & NIHR Biomedical Research Center at Guy's and St. Thomas' Hospital and King's College London, Guy's Hospital, King's College London , London , UK.,b Division of Cancer Studies, Faculty of Life Sciences and Medicine, King's College London, Guy's Hospital , London , UK
| | - Debra H Josephs
- a St. John's Institute of Dermatology, Division of Genetics and Molecular Medicine, Faculty of Life Sciences and Medicine, King's College London & NIHR Biomedical Research Center at Guy's and St. Thomas' Hospital and King's College London, Guy's Hospital, King's College London , London , UK.,b Division of Cancer Studies, Faculty of Life Sciences and Medicine, King's College London, Guy's Hospital , London , UK
| | - Giulia Pellizzari
- a St. John's Institute of Dermatology, Division of Genetics and Molecular Medicine, Faculty of Life Sciences and Medicine, King's College London & NIHR Biomedical Research Center at Guy's and St. Thomas' Hospital and King's College London, Guy's Hospital, King's College London , London , UK.,b Division of Cancer Studies, Faculty of Life Sciences and Medicine, King's College London, Guy's Hospital , London , UK
| | - James F Spicer
- b Division of Cancer Studies, Faculty of Life Sciences and Medicine, King's College London, Guy's Hospital , London , UK
| | - Ana Montes
- c Department of Medical Oncology , Guy's and St Thomas' NHS Foundation Trust , London , UK
| | - Sophia N Karagiannis
- a St. John's Institute of Dermatology, Division of Genetics and Molecular Medicine, Faculty of Life Sciences and Medicine, King's College London & NIHR Biomedical Research Center at Guy's and St. Thomas' Hospital and King's College London, Guy's Hospital, King's College London , London , UK
| |
Collapse
|
19
|
Ntanasis-Stathopoulos I, Fotopoulos G, Tzanninis IG, Kotteas EA. The Emerging Role of Tyrosine Kinase Inhibitors in Ovarian Cancer Treatment: A Systematic Review. Cancer Invest 2016; 34:313-39. [PMID: 27486869 DOI: 10.1080/07357907.2016.1206117] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The present systematic review summarizes current evidence regarding the mechanisms of action, the efficacy, and the adverse effects of tyrosine kinase inhibitors (TKIs) in ovarian cancer patients. Phase II and III clinical trials were sought in the PubMed database and in the Clinical Trials.gov registry through September 30, 2015. Seventy-five clinical trials regarding TKIs targeting mainly vascular endothelial growth factor receptor, epidermal growth factor receptor, platelet-derived growth factor receptor, and sarcoma tyrosine kinase (Src) were yielded. The most promising results were noted with cediranib, nintedanib, and pazopanib. However, drawing universal conclusions about the potential integration of TKIs in ovarian cancer therapy remains elusive. Furthermore, emerging challenges and directions for the future research are critically discussed.
Collapse
Affiliation(s)
| | - George Fotopoulos
- a Oncology Unit, Sotiria General Hospital , Athens School of Medicine , Athens , Greece
| | | | - Elias A Kotteas
- a Oncology Unit, Sotiria General Hospital , Athens School of Medicine , Athens , Greece
| |
Collapse
|
20
|
Expression and significance of Ku80 and PDGFR-α in nasal NK/T-cell lymphoma. Pathol Res Pract 2016; 212:204-9. [DOI: 10.1016/j.prp.2015.12.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 12/01/2015] [Accepted: 12/14/2015] [Indexed: 11/17/2022]
|
21
|
miR-140-5p inhibits ovarian cancer growth partially by repression of PDGFRA. Biomed Pharmacother 2015; 75:117-22. [PMID: 26297547 DOI: 10.1016/j.biopha.2015.07.035] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 07/26/2015] [Indexed: 12/15/2022] Open
Abstract
Dysregulation of miRNAs is a common feature in human cancers, and miR-140-5p has been found to be down-regulated in cancer. However its role in ovarian cancer remains unclear. miR-140-5p was underexpressed in HCC tissues and cell lines compared with their normal controls. Additionally, PDGFRA was predicted the target gene of miR-140-5p. PDGFRA was inversely correlated with the expression of miR-140-5p in ovarian cancer cells. Importantly, we demonstrate that the over expression of miR-140-5p significantly inhibits ovarian cancer cell proliferation and induces apoptosis. Our results suggest the existence of a novel miR-140-5p-PDGFRA pathway and indicate that miR-140-5p acts as a tumor suppressor during ovarian carcinogenesis. These results may provide a promising alterative strategy for the therapeutic treatment of ovarian cancer.
Collapse
|
22
|
Farooqi AA, Siddik ZH. Platelet-derived growth factor (PDGF) signalling in cancer: rapidly emerging signalling landscape. Cell Biochem Funct 2015; 33:257-65. [PMID: 26153649 DOI: 10.1002/cbf.3120] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 04/02/2015] [Accepted: 05/11/2015] [Indexed: 12/25/2022]
Abstract
Platelet-derived growth factor (PDGF)-mediated signalling has emerged as one of the most extensively and deeply studied biological mechanism reported to be involved in regulation of growth and survival of different cell types. However, overwhelmingly increasing scientific evidence is also emphasizing on dysregulation of spatio-temporally controlled PDGF-induced signalling as a basis for cancer development. We partition this multi-component review into recently developing understanding of dysregulation PDGF signalling in different cancers, how PDGF receptors are quantitatively controlled by microRNAs. Moreover, we also summarize most recent advancements in therapeutic targeting of PDGFR as evidenced by preclinical studies. Better understanding of the PDGF-induced intracellular signalling in different cancers will be helpful in catalysing the transition from a segmented view of cancer biology to a conceptual continuum.
Collapse
Affiliation(s)
| | - Zahid H Siddik
- University of Texas, MD Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
23
|
Arrondeau J, Huillard O, Tlemsani C, Cessot A, Boudou-Rouquette P, Blanchet B, Thomas-Schoemann A, Vidal M, Tigaud JM, Durand JP, Alexandre J, Goldwasser F. Investigational therapies up to Phase II which target PDGF receptors: potential anti-cancer therapeutics. Expert Opin Investig Drugs 2015; 24:673-87. [PMID: 25599887 DOI: 10.1517/13543784.2015.1005736] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION The platelet-derived growth factor receptor (PDGFR) pathway has important functions in cell growth and, by overexpression or mutation, could also be a driver for tumor development. Moreover, PDGFR is expressed in a tumoral microenvironment and could promote tumorigenesis. With these biological considerations, the PDGFR pathway could be an interesting target for therapeutics. Currently, there are many molecules under development that target the PDGFR pathway in different types of cancer. AREAS COVERED In this review, the authors report the different molecules under development, as well as those approved albeit briefly, which inhibit the PDGFR pathway. Furthermore, the authors summarize their specificities, their toxicities, and their development. EXPERT OPINION Currently, most PDGFR kinase inhibitors are multikinase inhibitors and therefore do not simply target the PDGFR pathway. The development of more specific PDGFR inhibitors could improve drug efficacy. Moreover, selecting tumors harboring mutations or amplifications of PDGFR could improve outcomes associated with the use of these molecules. The authors believe that new technologies, such as kinome arrays or pharmacologic assays, could be of benefit to understanding resistance mechanisms and develop more selective PDGFR inhibitors.
Collapse
Affiliation(s)
- Jennifer Arrondeau
- Paris Descartes University, Cochin Hospital, AP-HP, Medical Oncology Department, Angiogenesis Inhibitors Multidisciplinary Study Group (CERIA) , Paris , France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Kikuchi A, Monga SP. PDGFRα in liver pathophysiology: emerging roles in development, regeneration, fibrosis, and cancer. Gene Expr 2015; 16:109-27. [PMID: 25700367 PMCID: PMC4410163 DOI: 10.3727/105221615x14181438356210] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Platelet-derived growth factor receptor α (PDGFRα) is an isoform of the PDGFR family of tyrosine kinase receptors involved in cell proliferation, survival, differentiation, and growth. In this review, we highlight the role of PDGFRα and the current evidence of its expression and activities in liver development, regeneration, and pathology-including fibrosis, cirrhosis, and liver cancer. Studies elucidating PDGFRα signaling in processes ranging from profibrotic signaling, angiogenesis, and oxidative stress to epithelial-to-mesenchymal transition point toward PDGFRα as a potential therapeutic target in various hepatic pathologies, including hepatic fibrosis and liver cancer. Furthermore, PDGFRα localization and modulation during liver development and regeneration may lend insight into its potential roles in various pathologic states. We will also briefly discuss some of the current targeted treatments for PDGFRα, including multi receptor tyrosine kinase inhibitors and PDGFRα-specific inhibitors.
Collapse
Affiliation(s)
- Alexander Kikuchi
- Department of Pathology and Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | |
Collapse
|
25
|
Lin H, Lin D, Xiong XS, Dai XX, Lin T. Role of platelet-derived growth factor-α in eosinophilic and non-eosinophilic chronic rhinosinusitis with nasal polyps. Int Forum Allergy Rhinol 2014; 4:909-14. [PMID: 25256824 DOI: 10.1002/alr.21419] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Revised: 07/10/2014] [Accepted: 08/19/2014] [Indexed: 11/11/2022]
Abstract
BACKGROUND The pathogenesis of human chronic rhinosinusitis with nasal polyps (CRSwNP) comprising eosinophilic CRSwNP (ECRSwNP) and non-eosinophilic (nECRSwNP) is not completely understood. Recent evidence has suggested that platelet-derived growth factor receptor alpha (PDGFRα) is implicated in cell growth, transformation, proliferation, migration, and vascular permeability and platelet-derived growth factor-A (PDGF-A) is a specific ligand for PDGFRα. However, little is known about their roles in CRSwNP. Therefore, we aimed to investigate the expression and role of PDGFRα and PDGF-A in CRSwNP. METHODS PDGFRα protein expression was investigated by immunohistochemistry method and messenger RNA (mRNA) expression of PDGFRα and PDGF-A were assessed by real-time polymerase chain reaction (PCR) in CRSwNP patients and control subjects. Moreover, the effects of various stimulators with different concentrations and time on PDGFRα were evaluated on nasal explant culture. RESULTS Quantitative analysis of immunostaining for PDGFRα showed an obvious elevation in immunolabeling of PDGFRα in CRSwNP groups compared with controls. Furthermore, PDGFRα protein was significantly stronger expressed in ECRSwNP group than nECRSwNP group and atopic patients showed stronger expression of PDGFRα protein than nonatopic patients. The mRNA of PDGFRα and PDGF-A were overexpressed in CRSwNP, especially in ECRSwNP. PDGFRα mRNA expression was closely related to PDGF-A mRNA. In nasal explant culture and stimulation, PDGFRα mRNA was augmented by interleukin 4 (IL-4), IL-5, or IL-1β respectively, but suppressed by IL-27. CONCLUSION PDGFRα may play a pivotal role in the pathophysiology of ECRSwNP and nECRSwNP by combining with PDGF-A. IL-4, IL-5, or IL-1β may be critical for PDGFRα gene expression.
Collapse
Affiliation(s)
- Hai Lin
- Department of Otorhinolaryngology, Eye and ENT Hospital of Fudan University, Shanghai, China; Department of Otorhinolaryngology, Fuzhou General Hospital, Fuzhou, Fujian, China
| | | | | | | | | |
Collapse
|
26
|
Weigel MT, Rath K, Alkatout I, Wenners AS, Schem C, Maass N, Jonat W, Mundhenke C. Nilotinib in combination with carboplatin and paclitaxel is a candidate for ovarian cancer treatment. Oncology 2014; 87:232-45. [PMID: 25116401 DOI: 10.1159/000363656] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 04/28/2014] [Indexed: 11/19/2022]
Abstract
PURPOSE Nilotinib is a selective tyrosine kinase inhibitor of c-Kit, Abl and platelet-derived growth factor receptor-α/β. To evaluate nilotinib's potential use as a treatment of human ovarian cancer, we tested nilotinib's preclinical activity in ovarian cancer cell lines with different tyrosine kinase expression patterns. METHODS The effects of nilotinib on ovarian cancer cell growth were studied alone and in combination with carboplatin and paclitaxel. Proapoptotic and antimigratory effects were examined using TUNEL and migration assays. RESULTS Nilotinib alone and in combination with carboplatin and paclitaxel significantly inhibited cell growth in PDGFR-α-positive ovarian cancer cell lines. The combination of nilotinib with carboplatin and paclitaxel showed synergistic effects on cell proliferation. Nilotinib treatment led to the inhibition of cell migration alone and in combination with carboplatin and paclitaxel. Apoptosis induction occurred in response to nilotinib that increased in combination with carboplatin. CONCLUSIONS Nilotinib may be a feasible targeted therapy option for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Marion T Weigel
- Department of Gynecology and Obstetrics, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Farooqi AA, Yaylim I, Ozkan NE, Zaman F, Halim TA, Chang HW. Restoring TRAIL mediated signaling in ovarian cancer cells. Arch Immunol Ther Exp (Warsz) 2014; 62:459-74. [PMID: 25030086 DOI: 10.1007/s00005-014-0307-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 06/26/2014] [Indexed: 02/08/2023]
Abstract
Ovarian cancer has emerged as a multifaceted and genomically complex disease. Genetic/epigenetic mutations, suppression of tumor suppressors, overexpression of oncogenes, rewiring of intracellular signaling cascades and loss of apoptosis are some of the deeply studied mechanisms. In vitro and in vivo studies have highlighted different molecular mechanisms that regulate tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) mediated apoptosis in ovarian cancer. In this review, we bring to limelight, expansion in understanding systematical characterization of ovarian cancer cells has led to the rapid development of new drugs and treatments to target negative regulators of TRAIL mediated signaling pathway. Wide ranging synthetic and natural agents have been shown to stimulate mRNA and protein expression of death receptors. This review is compartmentalized into programmed cell death protein 4, platelet-derived growth factor signaling and miRNA control of TRAIL mediated signaling to ovarian cancer. Mapatumumab and PRO95780 have been tested for efficacy against ovarian cancer. Use of high-throughput screening assays will aid in dissecting the heterogeneity of this disease and increasing a long-term survival which might be achieved by translating rapidly accumulating information obtained from molecular and cellular studies to clinic researches.
Collapse
Affiliation(s)
- Ammad Ahmad Farooqi
- Laboratory for Translational Oncology and Personalized Medicine, RLMC, 35 km Ferozepur Road, Lahore, Pakistan,
| | | | | | | | | | | |
Collapse
|
28
|
Roh JW, Huang J, Hu W, Yang X, Jennings NB, Sehgal V, Sohn BH, Han HD, Lee SJ, Thanapprapasr D, Bottsford-Miller J, Zand B, Dalton HJ, Previs RA, Davis AN, Matsuo K, Lee JS, Ram P, Coleman RL, Sood AK. Biologic effects of platelet-derived growth factor receptor α blockade in uterine cancer. Clin Cancer Res 2014; 20:2740-50. [PMID: 24634380 PMCID: PMC4024372 DOI: 10.1158/1078-0432.ccr-13-2507] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE Platelet-derived growth factor receptor α (PDGFRα) expression is frequently observed in many kinds of cancer and is a candidate for therapeutic targeting. This preclinical study evaluated the biologic significance of PDGFRα and PDGFRα blockade (using a fully humanized monoclonal antibody, 3G3) in uterine cancer. EXPERIMENTAL DESIGN Expression of PDGFRα was examined in uterine cancer clinical samples and cell lines, and biologic effects of PDGFRα inhibition were evaluated using in vitro (cell viability, apoptosis, and invasion) and in vivo (orthotopic) models of uterine cancer. RESULTS PDGFRα was highly expressed and activated in uterine cancer samples and cell lines. Treatment with 3G3 resulted in substantial inhibition of PDGFRα phosphorylation and of downstream signaling molecules AKT and mitogen-activated protein kinase (MAPK). Cell viability and invasive potential of uterine cancer cells were also inhibited by 3G3 treatment. In orthotopic mouse models of uterine cancer, 3G3 monotherapy had significant antitumor effects in the PDGFRα-positive models (Hec-1A, Ishikawa, Spec-2) but not in the PDGFRα-negative model (OVCA432). Greater therapeutic effects were observed for 3G3 in combination with chemotherapy than for either drug alone in the PDGFRα-positive models. The antitumor effects of therapy were related to increased apoptosis and decreased proliferation and angiogenesis. CONCLUSIONS These findings identify PDGFRα as an attractive target for therapeutic development in uterine cancer.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Blotting, Western
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Drug Synergism
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Immunohistochemistry
- Mice, Nude
- Mitogen-Activated Protein Kinases/metabolism
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/prevention & control
- Oligonucleotide Array Sequence Analysis
- Phosphorylation/drug effects
- Proto-Oncogene Proteins c-akt/metabolism
- Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors
- Receptor, Platelet-Derived Growth Factor alpha/immunology
- Receptor, Platelet-Derived Growth Factor alpha/metabolism
- Signal Transduction/drug effects
- Transcriptome/drug effects
- Uterine Neoplasms/drug therapy
- Uterine Neoplasms/genetics
- Uterine Neoplasms/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Ju-Won Roh
- Authors' Affiliations: Departments of Gynecologic Oncology and Reproductive Medicine and Cancer Biology, Center for RNA Interference and Non-Coding RNA, the University of Texas MD Anderson Cancer Center, Houston, Texas; University of Southern California, Los Angeles, California; Department of Obstetrics and Gynecology, Dongguk University; Departments of Systems Biology and Obstetrics and Gynecology, Konkuk University, Seoul, Korea; and Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, ThailandAuthors' Affiliations: Departments of Gynecologic Oncology and Reproductive Medicine and Cancer Biology, Center for RNA Interference and Non-Coding RNA, the University of Texas MD Anderson Cancer Center, Houston, Texas; University of Southern California, Los Angeles, California; Department of Obstetrics and Gynecology, Dongguk University; Departments of Systems Biology and Obstetrics and Gynecology, Konkuk University, Seoul, Korea; and Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Jie Huang
- Authors' Affiliations: Departments of Gynecologic Oncology and Reproductive Medicine and Cancer Biology, Center for RNA Interference and Non-Coding RNA, the University of Texas MD Anderson Cancer Center, Houston, Texas; University of Southern California, Los Angeles, California; Department of Obstetrics and Gynecology, Dongguk University; Departments of Systems Biology and Obstetrics and Gynecology, Konkuk University, Seoul, Korea; and Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Wei Hu
- Authors' Affiliations: Departments of Gynecologic Oncology and Reproductive Medicine and Cancer Biology, Center for RNA Interference and Non-Coding RNA, the University of Texas MD Anderson Cancer Center, Houston, Texas; University of Southern California, Los Angeles, California; Department of Obstetrics and Gynecology, Dongguk University; Departments of Systems Biology and Obstetrics and Gynecology, Konkuk University, Seoul, Korea; and Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - XiaoYun Yang
- Authors' Affiliations: Departments of Gynecologic Oncology and Reproductive Medicine and Cancer Biology, Center for RNA Interference and Non-Coding RNA, the University of Texas MD Anderson Cancer Center, Houston, Texas; University of Southern California, Los Angeles, California; Department of Obstetrics and Gynecology, Dongguk University; Departments of Systems Biology and Obstetrics and Gynecology, Konkuk University, Seoul, Korea; and Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Nicholas B Jennings
- Authors' Affiliations: Departments of Gynecologic Oncology and Reproductive Medicine and Cancer Biology, Center for RNA Interference and Non-Coding RNA, the University of Texas MD Anderson Cancer Center, Houston, Texas; University of Southern California, Los Angeles, California; Department of Obstetrics and Gynecology, Dongguk University; Departments of Systems Biology and Obstetrics and Gynecology, Konkuk University, Seoul, Korea; and Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Vasudha Sehgal
- Authors' Affiliations: Departments of Gynecologic Oncology and Reproductive Medicine and Cancer Biology, Center for RNA Interference and Non-Coding RNA, the University of Texas MD Anderson Cancer Center, Houston, Texas; University of Southern California, Los Angeles, California; Department of Obstetrics and Gynecology, Dongguk University; Departments of Systems Biology and Obstetrics and Gynecology, Konkuk University, Seoul, Korea; and Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Bo Hwa Sohn
- Authors' Affiliations: Departments of Gynecologic Oncology and Reproductive Medicine and Cancer Biology, Center for RNA Interference and Non-Coding RNA, the University of Texas MD Anderson Cancer Center, Houston, Texas; University of Southern California, Los Angeles, California; Department of Obstetrics and Gynecology, Dongguk University; Departments of Systems Biology and Obstetrics and Gynecology, Konkuk University, Seoul, Korea; and Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Hee Dong Han
- Authors' Affiliations: Departments of Gynecologic Oncology and Reproductive Medicine and Cancer Biology, Center for RNA Interference and Non-Coding RNA, the University of Texas MD Anderson Cancer Center, Houston, Texas; University of Southern California, Los Angeles, California; Department of Obstetrics and Gynecology, Dongguk University; Departments of Systems Biology and Obstetrics and Gynecology, Konkuk University, Seoul, Korea; and Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Sun Joo Lee
- Authors' Affiliations: Departments of Gynecologic Oncology and Reproductive Medicine and Cancer Biology, Center for RNA Interference and Non-Coding RNA, the University of Texas MD Anderson Cancer Center, Houston, Texas; University of Southern California, Los Angeles, California; Department of Obstetrics and Gynecology, Dongguk University; Departments of Systems Biology and Obstetrics and Gynecology, Konkuk University, Seoul, Korea; and Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, ThailandAuthors' Affiliations: Departments of Gynecologic Oncology and Reproductive Medicine and Cancer Biology, Center for RNA Interference and Non-Coding RNA, the University of Texas MD Anderson Cancer Center, Houston, Texas; University of Southern California, Los Angeles, California; Department of Obstetrics and Gynecology, Dongguk University; Departments of Systems Biology and Obstetrics and Gynecology, Konkuk University, Seoul, Korea; and Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Duangmani Thanapprapasr
- Authors' Affiliations: Departments of Gynecologic Oncology and Reproductive Medicine and Cancer Biology, Center for RNA Interference and Non-Coding RNA, the University of Texas MD Anderson Cancer Center, Houston, Texas; University of Southern California, Los Angeles, California; Department of Obstetrics and Gynecology, Dongguk University; Departments of Systems Biology and Obstetrics and Gynecology, Konkuk University, Seoul, Korea; and Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, ThailandAuthors' Affiliations: Departments of Gynecologic Oncology and Reproductive Medicine and Cancer Biology, Center for RNA Interference and Non-Coding RNA, the University of Texas MD Anderson Cancer Center, Houston, Texas; University of Southern California, Los Angeles, California; Department of Obstetrics and Gynecology, Dongguk University; Departments of Systems Biology and Obstetrics and Gynecology, Konkuk University, Seoul, Korea; and Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Justin Bottsford-Miller
- Authors' Affiliations: Departments of Gynecologic Oncology and Reproductive Medicine and Cancer Biology, Center for RNA Interference and Non-Coding RNA, the University of Texas MD Anderson Cancer Center, Houston, Texas; University of Southern California, Los Angeles, California; Department of Obstetrics and Gynecology, Dongguk University; Departments of Systems Biology and Obstetrics and Gynecology, Konkuk University, Seoul, Korea; and Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Behrouz Zand
- Authors' Affiliations: Departments of Gynecologic Oncology and Reproductive Medicine and Cancer Biology, Center for RNA Interference and Non-Coding RNA, the University of Texas MD Anderson Cancer Center, Houston, Texas; University of Southern California, Los Angeles, California; Department of Obstetrics and Gynecology, Dongguk University; Departments of Systems Biology and Obstetrics and Gynecology, Konkuk University, Seoul, Korea; and Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Heather J Dalton
- Authors' Affiliations: Departments of Gynecologic Oncology and Reproductive Medicine and Cancer Biology, Center for RNA Interference and Non-Coding RNA, the University of Texas MD Anderson Cancer Center, Houston, Texas; University of Southern California, Los Angeles, California; Department of Obstetrics and Gynecology, Dongguk University; Departments of Systems Biology and Obstetrics and Gynecology, Konkuk University, Seoul, Korea; and Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Rebecca A Previs
- Authors' Affiliations: Departments of Gynecologic Oncology and Reproductive Medicine and Cancer Biology, Center for RNA Interference and Non-Coding RNA, the University of Texas MD Anderson Cancer Center, Houston, Texas; University of Southern California, Los Angeles, California; Department of Obstetrics and Gynecology, Dongguk University; Departments of Systems Biology and Obstetrics and Gynecology, Konkuk University, Seoul, Korea; and Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Ashley N Davis
- Authors' Affiliations: Departments of Gynecologic Oncology and Reproductive Medicine and Cancer Biology, Center for RNA Interference and Non-Coding RNA, the University of Texas MD Anderson Cancer Center, Houston, Texas; University of Southern California, Los Angeles, California; Department of Obstetrics and Gynecology, Dongguk University; Departments of Systems Biology and Obstetrics and Gynecology, Konkuk University, Seoul, Korea; and Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Koji Matsuo
- Authors' Affiliations: Departments of Gynecologic Oncology and Reproductive Medicine and Cancer Biology, Center for RNA Interference and Non-Coding RNA, the University of Texas MD Anderson Cancer Center, Houston, Texas; University of Southern California, Los Angeles, California; Department of Obstetrics and Gynecology, Dongguk University; Departments of Systems Biology and Obstetrics and Gynecology, Konkuk University, Seoul, Korea; and Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, ThailandAuthors' Affiliations: Departments of Gynecologic Oncology and Reproductive Medicine and Cancer Biology, Center for RNA Interference and Non-Coding RNA, the University of Texas MD Anderson Cancer Center, Houston, Texas; University of Southern California, Los Angeles, California; Department of Obstetrics and Gynecology, Dongguk University; Departments of Systems Biology and Obstetrics and Gynecology, Konkuk University, Seoul, Korea; and Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Ju-Seog Lee
- Authors' Affiliations: Departments of Gynecologic Oncology and Reproductive Medicine and Cancer Biology, Center for RNA Interference and Non-Coding RNA, the University of Texas MD Anderson Cancer Center, Houston, Texas; University of Southern California, Los Angeles, California; Department of Obstetrics and Gynecology, Dongguk University; Departments of Systems Biology and Obstetrics and Gynecology, Konkuk University, Seoul, Korea; and Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Prahlad Ram
- Authors' Affiliations: Departments of Gynecologic Oncology and Reproductive Medicine and Cancer Biology, Center for RNA Interference and Non-Coding RNA, the University of Texas MD Anderson Cancer Center, Houston, Texas; University of Southern California, Los Angeles, California; Department of Obstetrics and Gynecology, Dongguk University; Departments of Systems Biology and Obstetrics and Gynecology, Konkuk University, Seoul, Korea; and Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Robert L Coleman
- Authors' Affiliations: Departments of Gynecologic Oncology and Reproductive Medicine and Cancer Biology, Center for RNA Interference and Non-Coding RNA, the University of Texas MD Anderson Cancer Center, Houston, Texas; University of Southern California, Los Angeles, California; Department of Obstetrics and Gynecology, Dongguk University; Departments of Systems Biology and Obstetrics and Gynecology, Konkuk University, Seoul, Korea; and Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Anil K Sood
- Authors' Affiliations: Departments of Gynecologic Oncology and Reproductive Medicine and Cancer Biology, Center for RNA Interference and Non-Coding RNA, the University of Texas MD Anderson Cancer Center, Houston, Texas; University of Southern California, Los Angeles, California; Department of Obstetrics and Gynecology, Dongguk University; Departments of Systems Biology and Obstetrics and Gynecology, Konkuk University, Seoul, Korea; and Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, ThailandAuthors' Affiliations: Departments of Gynecologic Oncology and Reproductive Medicine and Cancer Biology, Center for RNA Interference and Non-Coding RNA, the University of Texas MD Anderson Cancer Center, Houston, Texas; University of Southern California, Los Angeles, California; Department of Obstetrics and Gynecology, Dongguk University; Departments of Systems Biology and Obstetrics and Gynecology, Konkuk University, Seoul, Korea; and Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, ThailandAuthors' Affiliations: Departments of Gynecologic Oncology and Reproductive Medicine and Cancer Biology, Center for RNA Interference and Non-Coding RNA, the University of Texas MD Anderson Cancer Center, Houston, Texas; University of Southern California, Los Angeles, California; Department of Obstetrics and Gynecology, Dongguk University; Departments of Systems Biology and Obstetrics and Gynecology, Konkuk University, Seoul, Korea; and Department of Obstetrics and Gynecology, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
29
|
Yang L, Moss T, Mangala LS, Marini J, Zhao H, Wahlig S, Armaiz-Pena G, Jiang D, Achreja A, Win J, Roopaimoole R, Rodriguez-Aguayo C, Mercado-Uribe I, Lopez-Berestein G, Liu J, Tsukamoto T, Sood AK, Ram PT, Nagrath D. Metabolic shifts toward glutamine regulate tumor growth, invasion and bioenergetics in ovarian cancer. Mol Syst Biol 2014; 10:728. [PMID: 24799285 PMCID: PMC4188042 DOI: 10.1002/msb.20134892] [Citation(s) in RCA: 246] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Glutamine can play a critical role in cellular growth in multiple cancers. Glutamine‐addicted cancer cells are dependent on glutamine for viability, and their metabolism is reprogrammed for glutamine utilization through the tricarboxylic acid (TCA) cycle. Here, we have uncovered a missing link between cancer invasiveness and glutamine dependence. Using isotope tracer and bioenergetic analysis, we found that low‐invasive ovarian cancer (OVCA) cells are glutamine independent, whereas high‐invasive OVCA cells are markedly glutamine dependent. Consistent with our findings, OVCA patients’ microarray data suggest that glutaminolysis correlates with poor survival. Notably, the ratio of gene expression associated with glutamine anabolism versus catabolism has emerged as a novel biomarker for patient prognosis. Significantly, we found that glutamine regulates the activation of STAT3, a mediator of signaling pathways which regulates cancer hallmarks in invasive OVCA cells. Our findings suggest that a combined approach of targeting high‐invasive OVCA cells by blocking glutamine's entry into the TCA cycle, along with targeting low‐invasive OVCA cells by inhibiting glutamine synthesis and STAT3 may lead to potential therapeutic approaches for treating OVCAs.
Collapse
Affiliation(s)
- Lifeng Yang
- Laboratory for Systems Biology of Human Diseases, Rice University, Houston, TX, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Growing understanding of the role of thrombocytosis, high platelet turnover, and the presence of activated platelets in the circulation in cancer progression and metastasis has brought megakaryocytes into focus. Platelet biology is essential to hemostasis, vascular integrity, angiogenesis, inflammation, innate immunity, wound healing, and cancer biology. However, before megakaryocyte/platelet-directed therapies can be considered for clinical use, understanding of the mechanism and biology of paraneoplastic thrombocytosis in malignancy is required. Here, we provide an overview of the clinical implications, biological significance, and mechanisms of paraneoplastic thrombocytosis in the context of ovarian cancer.
Collapse
Affiliation(s)
- Ashley N Davis
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Vahid Afshar-Kharghan
- Department of Benign Hematology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX; Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TTX; Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|