1
|
Yue SY, Niu D, Ma WM, Guan Y, Liu QS, Wang XB, Xiao YZ, Meng J, Ding K, Zhang L, Du HX, Liang CZ. The CXCL10/CXCR3 axis regulates Th1 cell differentiation and migration in experimental autoimmune prostatitis through the PI3K/AKT pathway. Andrology 2024; 12:1408-1418. [PMID: 38095276 DOI: 10.1111/andr.13571] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 10/23/2023] [Accepted: 11/23/2023] [Indexed: 08/15/2024]
Abstract
OBJECTIVE To investigate the mechanism of the CXCL10/CXCR3 axis regulating Th1 cell differentiation and migration through the PI3K/AKT pathway in chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS). METHODS Experimental autoimmune prostatitis (EAP) model, a well-described and validated animal model of CP/CPPS, was used in our study. After treatment with CXCL10, the severity of EAP and Th1 cell proportion were respectively measured by HE stains, immunohistochemistry, and flow cytometry. Then, the protein expression of the PI3K/AKT pathway in CXCL10/CXCR3-regulated Th1 cell differentiation and migration was evaluated by western blotting. Additionally, by the CXCR3 antagonist AMG487 and the PI3K inhibitor LY294002 applications, the effects of CXCL10/CXCR3 through PI3K/AKT pathway on the Th1 cell differentiation and migration were further assessed. RESULTS The EAP model was successfully built. CXCL10 increased the proportion of Th1 cells in EAP mice, accompanied by upregulation of the PI3K/AKT pathway. Additionally, the PI3K/AKT pathway was found to be involved in CXCL10/CXCR3 axis-mediated Th1 cell differentiation and migration. CONCLUSIONS Our investigations indicate that the CXCL10/CXCR3 axis regulates Th1 cell differentiation and migration in EAP through the PI3K/AKT pathway, which provides a new perspective on the immunological mechanisms of CP/CPPS.
Collapse
Affiliation(s)
- Shao-Yu Yue
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Di Niu
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Wen-Ming Ma
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yu Guan
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Qiu-Shi Liu
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Xiao-Bin Wang
- Department of Urology, Southern University of Science and Technology Hospital, Shenzhen, China
| | - Yun-Zheng Xiao
- Department of Urology, Southern University of Science and Technology Hospital, Shenzhen, China
| | - Jialin Meng
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Ke Ding
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Li Zhang
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - He-Xi Du
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Chao-Zhao Liang
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
2
|
Choi Y, Park NJY, Le TM, Lee E, Lee D, Nguyen HDT, Cho J, Park JY, Han HS, Chong GO. Immune Pathway and Gene Database (IMPAGT) Revealed the Immune Dysregulation Dynamics and Overactivation of the PI3K/Akt Pathway in Tumor Buddings of Cervical Cancer. Curr Issues Mol Biol 2022; 44:5139-5152. [PMID: 36354662 PMCID: PMC9688570 DOI: 10.3390/cimb44110350] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/17/2022] [Accepted: 10/21/2022] [Indexed: 08/31/2023] Open
Abstract
Tumor budding (TB) is a small cluster of malignant cells at the invasive front of a tumor. Despite being an adverse prognosis marker, little research has been conducted on the tumor immune microenvironment of tumor buddings, especially in cervical cancer. Therefore, RNA sequencing was performed using 21 formalin-fixed, paraffin-embedded slides of cervical tissues, and differentially expressed genes (DEGs) were analyzed. Immune Pathway and Gene Database (IMPAGT) was generated for immune profiling. "Pathway in Cancer" was identified as the most enriched pathway for both up- and downregulated DEGs. Kyoto Encyclopedia of Genes and Genomes Mapper and Gene Ontology further revealed the activation of the PI3K/Akt signaling pathway. An IMPAGT analysis revealed immune dysregulation even at the tumor budding stage, especially in the PI3K/Akt/mTOR axis, with a high efficiency and integrity. These findings emphasized the clinical significance of tumor buddings and the necessity of blocking the overactivation of the PI3K/Akt/mTOR pathway to improve targeted therapy in cervical cancer.
Collapse
Affiliation(s)
- Yeseul Choi
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Korea
- BK21 Four Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Nora Jee-Young Park
- Department of Pathology, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Department of Pathology, Kyungpook National University Chilgok Hospital, Daegu 41404, Korea
- Clinical Omics Institute, Kyungpook National University, Daegu 41405, Korea
| | - Tan Minh Le
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Korea
- BK21 Four Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Eunmi Lee
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Korea
- BK21 Four Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Donghyeon Lee
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Korea
- BK21 Four Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Hong Duc Thi Nguyen
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Korea
- BK21 Four Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Junghwan Cho
- Clinical Omics Institute, Kyungpook National University, Daegu 41405, Korea
| | - Ji-Young Park
- Department of Pathology, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Department of Pathology, Kyungpook National University Chilgok Hospital, Daegu 41404, Korea
| | - Hyung Soo Han
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Korea
- BK21 Four Program, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Clinical Omics Institute, Kyungpook National University, Daegu 41405, Korea
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Gun Oh Chong
- Clinical Omics Institute, Kyungpook National University, Daegu 41405, Korea
- Department of Obstetrics and Gynecology, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Department of Obstetrics and Gynecology, Kyungpook National University Chilgok Hospital, Daegu 41404, Korea
| |
Collapse
|
3
|
Wong OGW, Li J, Cheung ANY. Targeting DNA Damage Response Pathway in Ovarian Clear Cell Carcinoma. Front Oncol 2021; 11:666815. [PMID: 34737943 PMCID: PMC8560708 DOI: 10.3389/fonc.2021.666815] [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: 02/11/2021] [Accepted: 09/08/2021] [Indexed: 12/24/2022] Open
Abstract
Ovarian clear cell carcinoma (OCCC) is one of the major types of ovarian cancer and is of higher relative prevalence in Asians. It also shows higher possibility of resistance to cisplatin-based chemotherapy leading to poor prognosis. This may be attributed to the relative lack of mutations and aberrations in homologous recombination-associated genes, which are crucial in DNA damage response (DDR), such as BRCA1, BRCA2, p53, RAD51, and genes in the Fanconi anemia pathway. On the other hand, OCCC is characterized by a number of genetic defects rendering it vulnerable to DDR-targeting therapy, which is emerging as a potent treatment strategy for various cancer types. Mutations of ARID1A, PIK3CA, PTEN, and catenin beta 1 (CTNNB1), as well as overexpression of transcription factor hepatocyte nuclear factor-1β (HNF-1β), and microsatellite instability are common in OCCC. Of particular note is the loss-of-function mutations in ARID1A, which is found in approximately 50% of OCCC. ARID1A is crucial for processing of DNA double-strand break (DSB) and for sustaining DNA damage signaling, rendering ARID1A-deficient cells prone to impaired DNA damage checkpoint regulation and hence sensitive to poly ADP ribose polymerase (PARP) inhibitors. However, while preclinical studies have demonstrated the possibility to exploit DDR deficiency in OCCC for therapeutic purpose, progress in clinical application is lagging. In this review, we will recapitulate the preclinical studies supporting the potential of DDR targeting in OCCC treatment, with emphasis on the role of ARID1A in DDR. Companion diagnostic tests (CDx) for predicting susceptibility to PARP inhibitors are rapidly being developed for solid tumors including ovarian cancers and may readily be applicable on OCCC. The potential of various available DDR-targeting drugs for treating OCCC by drawing analogies with other solid tumors sharing similar genetic characteristics with OCCC will also be discussed.
Collapse
|
4
|
Jiang N, Dai Q, Su X, Fu J, Feng X, Peng J. Role of PI3K/AKT pathway in cancer: the framework of malignant behavior. Mol Biol Rep 2020; 47:4587-4629. [PMID: 32333246 PMCID: PMC7295848 DOI: 10.1007/s11033-020-05435-1] [Citation(s) in RCA: 373] [Impact Index Per Article: 74.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/03/2020] [Indexed: 12/12/2022]
Abstract
Given that the PI3K/AKT pathway has manifested its compelling influence on multiple cellular process, we further review the roles of hyperactivation of PI3K/AKT pathway in various human cancers. We state the abnormalities of PI3K/AKT pathway in different cancers, which are closely related with tumorigenesis, proliferation, growth, apoptosis, invasion, metastasis, epithelial-mesenchymal transition, stem-like phenotype, immune microenvironment and drug resistance of cancer cells. In addition, we investigated the current clinical trials of inhibitors against PI3K/AKT pathway in cancers and found that the clinical efficacy of these inhibitors as monotherapy has so far been limited despite of the promising preclinical activity, which means combinations of targeted therapy may achieve better efficacies in cancers. In short, we hope to feature PI3K/AKT pathway in cancers to the clinic and bring the new promising to patients for targeted therapies.
Collapse
Affiliation(s)
- Ningni Jiang
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Guangzhou, 510150 China
- The Third Clinical School of Guangzhou Medical University, Guangzhou, 510150 China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, 510150 China
| | - Qijie Dai
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Guangzhou, 510150 China
- The Third Clinical School of Guangzhou Medical University, Guangzhou, 510150 China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, 510150 China
| | - Xiaorui Su
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Guangzhou, 510150 China
- The Third Clinical School of Guangzhou Medical University, Guangzhou, 510150 China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, 510150 China
| | - Jianjiang Fu
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Guangzhou, 510150 China
- The Third Clinical School of Guangzhou Medical University, Guangzhou, 510150 China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, 510150 China
| | - Xuancheng Feng
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Guangzhou, 510150 China
- The Third Clinical School of Guangzhou Medical University, Guangzhou, 510150 China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, 510150 China
| | - Juan Peng
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Guangzhou, 510150 China
- The Third Clinical School of Guangzhou Medical University, Guangzhou, 510150 China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, 510150 China
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| |
Collapse
|
5
|
Im H, Lee J, Ryu KY, Yi JY. Integrin αvβ3-Akt signalling plays a role in radioresistance of melanoma. Exp Dermatol 2020; 29:562-569. [PMID: 32298492 DOI: 10.1111/exd.14102] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 02/07/2023]
Abstract
Melanoma is a deadly type of skin cancer that is particularly difficult to treat owing to its resistance to radiation therapy. Here, we attempted to determine the key proteins responsible for melanoma radioresistance, with the aim of improving disease response to radiation therapy. Two melanoma cell lines, SK-Mel5 and SK-Mel28, with different radiosensitivities were analysed via RNA-Seq (Quant-Seq) and target proteins with higher abundance in the more radioresistant cell line, SK-Mel28, identified. Among these proteins, integrin αvβ3, a well-known molecule in cell adhesion, was selected for analysis. Treatment of SK-Mel28 cells with cilengitide, an integrin αvβ3 inhibitor, as well as γ-irradiation resulted in more significant cell death than γ-irradiation alone. In addition, Akt, a downstream signal transducer of integrin αvβ3, showed high basic activation in SK-Mel28 and was significantly decreased upon co-treatment with cilengitide and γ-irradiation. MK-2206, an Akt inhibitor, exerted similar effects on the SK-Mel28 cell line following γ-irradiation. Our results collectively demonstrate that the integrin αvβ3-Akt signalling pathway contributes to radioresistance in SK-Mel28 cells, which may be manipulated to improve therapeutic options for melanoma.
Collapse
Affiliation(s)
- Hyuntaik Im
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, Korea.,Department of Life Science, University of Seoul, Seoul, Korea
| | - Jeeyong Lee
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Kwon-Yul Ryu
- Department of Life Science, University of Seoul, Seoul, Korea
| | - Jae Youn Yi
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| |
Collapse
|
6
|
Iida M, Harari PM, Wheeler DL, Toulany M. Targeting AKT/PKB to improve treatment outcomes for solid tumors. Mutat Res 2020; 819-820:111690. [PMID: 32120136 DOI: 10.1016/j.mrfmmm.2020.111690] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/31/2020] [Accepted: 02/11/2020] [Indexed: 12/16/2022]
Abstract
The serine/threonine kinase AKT, also known as protein kinase B (PKB), is the major substrate to phosphoinositide 3-kinase (PI3K) and consists of three paralogs: AKT1 (PKBα), AKT2 (PKBβ) and AKT3 (PKBγ). The PI3K/AKT pathway is normally activated by binding of ligands to membrane-bound receptor tyrosine kinases (RTKs) as well as downstream to G-protein coupled receptors and integrin-linked kinase. Through multiple downstream substrates, activated AKT controls a wide variety of cellular functions including cell proliferation, survival, metabolism, and angiogenesis in both normal and malignant cells. In human cancers, the PI3K/AKT pathway is most frequently hyperactivated due to mutations and/or overexpression of upstream components. Aberrant expression of RTKs, gain of function mutations in PIK3CA, RAS, PDPK1, and AKT itself, as well as loss of function mutation in AKT phosphatases are genetic lesions that confer hyperactivation of AKT. Activated AKT stimulates DNA repair, e.g. double strand break repair after radiotherapy. Likewise, AKT attenuates chemotherapy-induced apoptosis. These observations suggest that a crucial link exists between AKT and DNA damage. Thus, AKT could be a major predictive marker of conventional cancer therapy, molecularly targeted therapy, and immunotherapy for solid tumors. In this review, we summarize the current understanding by which activated AKT mediates resistance to cancer treatment modalities, i.e. radiotherapy, chemotherapy, and RTK targeted therapy. Next, the effect of AKT on response of tumor cells to RTK targeted strategies will be discussed. Finally, we will provide a brief summary on the clinical trials of AKT inhibitors in combination with radiochemotherapy, RTK targeted therapy, and immunotherapy.
Collapse
Affiliation(s)
- M Iida
- Department of Human Oncology, University of Wisconsin in Madison, Madison, WI, USA.
| | - P M Harari
- Department of Human Oncology, University of Wisconsin in Madison, Madison, WI, USA
| | - D L Wheeler
- Department of Human Oncology, University of Wisconsin in Madison, Madison, WI, USA
| | - M Toulany
- Division of Radiobiology and Molecular Environmental Research, Department of Radiation Oncology, University of Tuebingen, Tuebingen, Germany; German Cancer Consortium (DKTK), Partner Site Tuebingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
7
|
Jiang W, Wu Y, He T, Zhu H, Ke G, Xiang L, Yang H. Targeting of β-Catenin Reverses Radioresistance of Cervical Cancer with the PIK3CA-E545K Mutation. Mol Cancer Ther 2019; 19:337-347. [PMID: 31666350 DOI: 10.1158/1535-7163.mct-19-0309] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 09/11/2019] [Accepted: 10/22/2019] [Indexed: 11/16/2022]
Abstract
This study aims to explore whether E545K, the most common hotspot mutation of PIK3CA in cervical cancer, confers radioresistance to cervical cancer cells, to demonstrate the underling mechanism, and to develop the effective targets. SiHa and MS751 cells with PIK3CA-WT and PIK3CA-E545K were established by lentiviral transfection. The radiosensitivity was assessed by colony formation, cell cycle, cell apoptosis, DNA damage, and repair assay. The growth and immunohistochemical assay of xenograft tumor-related toxicity were evaluated in vivo It was indicated that more cells with PIK3CA-E545K arrested in S phase. Irradiation (IR) led to more survival percentage, less apoptosis, fewer pH2A.X foci, and higher expression of Chk1/2 in SiHa and MS751 cells bearing PIK3CA-E545K. Mechanically, AKT/GSK3β/β-catenin pathway was highly activated, and more β-catenin was found accumulated in nucleus in cells with PIK3CA-E545K after IR. Furthermore, targeting β-catenin by shRNA or XAV939 enhanced IR sensitivity in cells with PIK3CA-WT and PIK3CA-E545K, whereas it was more notably in the latter. β-Catenin shRNA and XAV939 increased IR-mediated inhibition of colony formation with highly activated p53/bcl2/bax pathway. XAV939 enhanced IR-caused apoptosis, DNA damage, overcame S-phase arrest, DNA repair and reversed β-catenin nuclear accumulation in MS751 cells with PIK3CA-E545K. In vivo, XAV939 enhanced the radiosensitivity of cervical cancer xenografts with PIK3CA-E545K with invisible viscera toxicity. The findings demonstrate that cervical cancer cells with PIK3CA-E545K are resistant to IR by enhancing the expression and nuclear accumulation of β-catenin. Targeting β-catenin reverses the radioresistance, which suggests possible areas for preclinical research on β-catenin inhibition for strengthening the radiosensitivity of cervical cancer.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yutuan Wu
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tiancong He
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hanting Zhu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Guihao Ke
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Libing Xiang
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Huijuan Yang
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
8
|
Toulany M. Targeting DNA Double-Strand Break Repair Pathways to Improve Radiotherapy Response. Genes (Basel) 2019; 10:genes10010025. [PMID: 30621219 PMCID: PMC6356315 DOI: 10.3390/genes10010025] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 12/07/2018] [Accepted: 12/27/2018] [Indexed: 12/13/2022] Open
Abstract
More than half of cancer patients receive radiotherapy as a part of their cancer treatment. DNA double-strand breaks (DSBs) are considered as the most lethal form of DNA damage and a primary cause of cell death and are induced by ionizing radiation (IR) during radiotherapy. Many malignant cells carry multiple genetic and epigenetic aberrations that may interfere with essential DSB repair pathways. Additionally, exposure to IR induces the activation of a multicomponent signal transduction network known as DNA damage response (DDR). DDR initiates cell cycle checkpoints and induces DSB repair in the nucleus by non-homologous end joining (NHEJ) or homologous recombination (HR). The canonical DSB repair pathways function in both normal and tumor cells. Thus, normal-tissue toxicity may limit the targeting of the components of these two pathways as a therapeutic approach in combination with radiotherapy. The DSB repair pathways are also stimulated through cytoplasmic signaling pathways. These signaling cascades are often upregulated in tumor cells harboring mutations or the overexpression of certain cellular oncogenes, e.g., receptor tyrosine kinases, PIK3CA and RAS. Targeting such cytoplasmic signaling pathways seems to be a more specific approach to blocking DSB repair in tumor cells. In this review, a brief overview of cytoplasmic signaling pathways that have been reported to stimulate DSB repair is provided. The state of the art of targeting these pathways will be discussed. A greater understanding of the underlying signaling pathways involved in DSB repair may provide valuable insights that will help to design new strategies to improve treatment outcomes in combination with radiotherapy.
Collapse
Affiliation(s)
- Mahmoud Toulany
- Division of Radiobiology and Molecular Environmental Research, Department of Radiation Oncology, University of Tuebingen, Roentgenweg 11, 72076 Tuebingen, Germany.
| |
Collapse
|
9
|
Lachkar B, Minaguchi T, Akiyama A, Liu S, Zhang S, Xu C, Shikama A, Tasaka N, Sakurai M, Nakao S, Ochi H, Yoshikawa H, Satoh T. Prognostic significance of PIK3CA mutation in stage IIB to IVA cervical cancers treated by concurrent chemoradiotherapy with weekly cisplatin. Medicine (Baltimore) 2018; 97:e11392. [PMID: 30075505 PMCID: PMC6081058 DOI: 10.1097/md.0000000000011392] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The standard treatment for locally advanced cervical cancer is cisplatin-based concurrent chemoradiotherapy (CCRT). Although the activated PI3-kinase/Akt pathway is known to be involved in both cisplatin-resistance and radioresistance, to date, only a few studies have reported significant associations between PIK3CA gene mutational status and outcome by CCRT in the disease. The aim of this study was to clarify the prognostic significance of PIK3CA mutational status in cervical cancers treated by CCRT.We analyzed PIK3CA mutation in 59 patients with stage IIB to IVA cervical carcinomas primarily treated by CCRT with weekly cisplatin using formalin-fixed paraffin-embedded biopsy specimens before treatment. Fifty-seven of 59 patients (97%) had locally advanced cancers with stage IIIA to IVA. Clinicopathologic data and patient survival were retrospectively compared according to PIK3CA mutational status.PIK3CA mutation was found in 7 of 59 patients (12%). No significant differences in clinicopathologic characteristics were observed according to PIK3CA mutational status. Patients with wild-type PIK3CA showed significantly improved cancer-specific survival as compared with mutated patients (P = .044). Subsequent survival analyses revealed that PIK3CA mutation was a significant prognostic factor for poor overall survival [multivariate adjusted hazard ratio (HR), 3.9; 95% confidence interval (95% CI), 1.3-11.8; P = .017] and cancer-specific survival (multivariate adjusted HR, 3.6; 95% CI, 1.2-11.0; P = .024).Together with previous published findings, the current study further supports the clinical significance of PIK3CA mutation in cervical cancer. Our observations suggest that molecular inhibitors targeting the PI3-kinase/Akt pathway may improve the outcome by CCRT in cervical cancers harboring PIK3CA mutation, providing significant implications for novel treatment strategy based on precision medicine in the disease.
Collapse
Affiliation(s)
- Bouchra Lachkar
- Doctoral Program in Obstetrics and Gynecology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba
| | - Takeo Minaguchi
- Department of Obstetrics and Gynecology, Faculty of Medicine
| | - Azusa Akiyama
- Department of Obstetrics and Gynecology, Faculty of Medicine
| | - Shuling Liu
- Doctoral Program in Obstetrics and Gynecology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba
| | - Shuang Zhang
- Doctoral Program in Obstetrics and Gynecology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba
| | - Chenyang Xu
- Doctoral Program in Obstetrics and Gynecology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba
| | - Ayumi Shikama
- Department of Obstetrics and Gynecology, Faculty of Medicine
| | - Nobutaka Tasaka
- Department of Obstetrics and Gynecology, Faculty of Medicine
| | - Manabu Sakurai
- Department of Obstetrics and Gynecology, Faculty of Medicine
| | - Sari Nakao
- Department of Obstetrics and Gynecology, Faculty of Medicine
| | - Hiroyuki Ochi
- Department of Obstetrics and Gynecology, Faculty of Medicine
| | - Hiroyuki Yoshikawa
- Department of Obstetrics and Gynecology, Faculty of Medicine
- Ibaraki Prefectural Central Hospital, Ibaraki, Japan
| | - Toyomi Satoh
- Department of Obstetrics and Gynecology, Faculty of Medicine
| |
Collapse
|
10
|
Park JH, Jung KH, Kim SJ, Fang Z, Yan HH, Son MK, Kim J, Kang YW, Lee JE, Han B, Lim JH, Hong SS. Radiosensitization of the PI3K inhibitor HS-173 through reduction of DNA damage repair in pancreatic cancer. Oncotarget 2017; 8:112893-112906. [PMID: 29348875 PMCID: PMC5762560 DOI: 10.18632/oncotarget.22850] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 11/11/2017] [Indexed: 01/05/2023] Open
Abstract
Activation of PI3K/AKT pathway occurs frequently in tumors and is correlated with radioresistance. The PI3K/AKT pathway can be an important target for improvement of radiotherapy. Although adding of chemotherapy to radiation therapy regimen enhances survival in patients with locally advanced pancreatic cancer, more effective therapies for increasing radiosensitivity are urgently needed. In this study, we investigated whether the novel PI3K inhibitor HS-173 could attenuate radiation-induced up-regulation of DNA damage repair processes and assessed its efficacy as a radio- and chemo-sensitizer. Radiosensitizing effects of HS-173 were tested in human pancreatic cells using clonogenic survival and growth assays. Mechanisms underlying the effects of HS-173 and radiation were determined by assessing cell cycle and DNA damage- repair pathway components, including ataxia-telangiectasia mutated (ATM) and DNA-dependent protein kinase catalytic subunit (DNA-PKcs). The in vivo efficacy of HS-173 in cancer radiotherapy was evaluated using a human tumor xenograft model. HS-173 significantly increased the sensitivity of pancreatic cancer cells to radiation, an effect that was associated with G2/M cell cycle arrest. HS-173 also significantly attenuated DNA damage repair by potently inhibiting ATM and DNA-PKcs, the two major kinases that respond to radiation-induced DNA double-strand breaks (DSBs), resulting in sustained DNA damage. Moreover, the combination of HS-173 and radiation delayed tumor growth and impaired DNA repair in a pancreatic cancer xenograft model, reflecting enhanced radiosensitization. These results showed that HS-173 significantly improved radiotherapy by inhibiting the DNA damage-repair pathway in pancreatic cancer. We therefore suggest that HS-173 may be an effective radiosensitizer for pancreatic cancer.
Collapse
Affiliation(s)
- Jung Hee Park
- Department of Drug Development, College of Medicine, Inha University, Sinheung-dong, Jung-gu, Incheon 400-712, Republic of Korea
| | - Kyung Hee Jung
- Department of Drug Development, College of Medicine, Inha University, Sinheung-dong, Jung-gu, Incheon 400-712, Republic of Korea
| | - Soo Jung Kim
- Department of Drug Development, College of Medicine, Inha University, Sinheung-dong, Jung-gu, Incheon 400-712, Republic of Korea
| | - Zhenghuan Fang
- Department of Drug Development, College of Medicine, Inha University, Sinheung-dong, Jung-gu, Incheon 400-712, Republic of Korea
| | - Hong Hua Yan
- Department of Drug Development, College of Medicine, Inha University, Sinheung-dong, Jung-gu, Incheon 400-712, Republic of Korea
| | - Mi Kwon Son
- Department of Drug Development, College of Medicine, Inha University, Sinheung-dong, Jung-gu, Incheon 400-712, Republic of Korea
| | - Juyoung Kim
- Department of Drug Development, College of Medicine, Inha University, Sinheung-dong, Jung-gu, Incheon 400-712, Republic of Korea
| | - Yeo Wool Kang
- Department of Drug Development, College of Medicine, Inha University, Sinheung-dong, Jung-gu, Incheon 400-712, Republic of Korea
| | - Ji Eun Lee
- Department of Drug Development, College of Medicine, Inha University, Sinheung-dong, Jung-gu, Incheon 400-712, Republic of Korea
| | - Boreum Han
- Department of Drug Development, College of Medicine, Inha University, Sinheung-dong, Jung-gu, Incheon 400-712, Republic of Korea
| | - Joo Han Lim
- Department of Internal Medicine, College of Medicine, Inha University, Sinheung-dong, Jung-gu, Incheon 400-712, Republic of Korea
| | - Soon-Sun Hong
- Department of Drug Development, College of Medicine, Inha University, Sinheung-dong, Jung-gu, Incheon 400-712, Republic of Korea
| |
Collapse
|
11
|
Activating Akt1 mutations alter DNA double strand break repair and radiosensitivity. Sci Rep 2017; 7:42700. [PMID: 28209968 PMCID: PMC5314324 DOI: 10.1038/srep42700] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 01/12/2017] [Indexed: 12/19/2022] Open
Abstract
The survival kinase Akt has clinical relevance to radioresistance. However, its contributions to the DNA damage response, DNA double strand break (DSB) repair and apoptosis remain poorly defined and often contradictory. We used a genetic approach to explore the consequences of genetic alterations of Akt1 for the cellular radiation response. While two activation-associated mutants with prominent nuclear access, the phospho-mimicking Akt1-TDSD and the clinically relevant PH-domain mutation Akt1-E17K, accelerated DSB repair and improved survival of irradiated Tramp-C1 murine prostate cancer cells and Akt1-knockout murine embryonic fibroblasts in vitro, the classical constitutively active membrane-targeted myrAkt1 mutant had the opposite effects. Interestingly, DNA-PKcs directly phosphorylated Akt1 at S473 in an in vitro kinase assay but not vice-versa. Pharmacological inhibition of DNA-PKcs or Akt restored radiosensitivity in tumour cells expressing Akt1-E17K or Akt1-TDSD. In conclusion, Akt1-mediated radioresistance depends on its activation state and nuclear localization and is accessible to pharmacologic inhibition.
Collapse
|
12
|
Gandhi AK. Novel agents and treatment techniques to enhance radiotherapeutic outcomes in carcinoma of the uterine cervix. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:49. [PMID: 26904571 DOI: 10.3978/j.issn.2305-5839.2015.10.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
BACKGROUND Survival of patients with locally advanced carcinoma cervix (LACC) using the current standard of concurrent chemo-radiotherapy (CCRT) has reached a plateau over the last two decades. Loco-regional failure in first two years of treatment completion and distant metastasis in the subsequent years has put the survival curves at a halt. Strategies of induction and adjuvant chemotherapy have yielded little as has any advancement in techniques of delivery of radiation therapy. This article aims at discussing the current existing literature as well as promising novel strategies to enhance radiotherapeutic outcomes in carcinoma of the uterine cervix. METHODS The review of English literature included phase I-III trials evaluating either a novel agent, novel application/modifications of an existing treatment regimen or an innovative treatment technique. The studies have been divided in to subsections with summary of most important findings at the end of each section. RESULTS Despite CCRT being the 'gold standard' treatment, several issues like optimum drug combination, schedule of drug delivery, combination with molecular targeted agents etc. remain undefined. Taxane, topoisomerase and gemcitabine based regimen needs to be further explored and compared with cisplatin based CCRT regimen. Several approaches like local delivery of cytotoxic agents, use of nano-medicine with CCRT are appearing on horizon with promises for the future. Therapies need to be designed based on the human papillomavirus titers of the patients and incorporation of radiosensitizers as an effective way of palliation with short course of radiotherapy may further enhance the radiotherapeutic outcomes. CONCLUSIONS The results of the studies with novel agents and treatment techniques appear promising. Further research in this arena including incorporation of cost-effectiveness analysis and quality of life issues in future trial designs are warranted.
Collapse
Affiliation(s)
- Ajeet Kumar Gandhi
- Department of Radiation Oncology, All India Institute of Medical Sciences, New Delhi 110029, India
| |
Collapse
|
13
|
Dolman MEM, van der Ploeg I, Koster J, Bate-Eya LT, Versteeg R, Caron HN, Molenaar JJ. DNA-Dependent Protein Kinase As Molecular Target for Radiosensitization of Neuroblastoma Cells. PLoS One 2015; 10:e0145744. [PMID: 26716839 PMCID: PMC4696738 DOI: 10.1371/journal.pone.0145744] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 12/08/2015] [Indexed: 11/18/2022] Open
Abstract
Tumor cells might resist therapy with ionizing radiation (IR) by non-homologous end-joining (NHEJ) of IR-induced double-strand breaks. One of the key players in NHEJ is DNA-dependent protein kinase (DNA-PK). The catalytic subunit of DNA-PK, i.e. DNA-PKcs, can be inhibited with the small-molecule inhibitor NU7026. In the current study, the in vitro potential of NU7026 to radiosensitize neuroblastoma cells was investigated. DNA-PKcs is encoded by the PRKDC (protein kinase, DNA-activated, catalytic polypeptide) gene. We showed that PRKDC levels were enhanced in neuroblastoma patients and correlated with a more advanced tumor stage and poor prognosis, making DNA-PKcs an interesting target for radiosensitization of neuroblastoma tumors. Optimal dose finding for combination treatment with NU7026 and IR was performed using NGP cells. One hour pre-treatment with 10 μM NU7026 synergistically sensitized NGP cells to 0.63 Gy IR. Radiosensitizing effects of NU7026 increased in time, with maximum effects observed from 96 h after IR-exposure on. Combined treatment of NGP cells with 10 μM NU7026 and 0.63 Gy IR resulted in apoptosis, while no apoptotic response was observed for either of the therapies alone. Inhibition of IR-induced DNA-PK activation by NU7026 confirmed the capability of NGP cells to, at least partially, resist IR by NHEJ. NU7026 also synergistically radiosensitized other neuroblastoma cell lines, while no synergistic effect was observed for low DNA-PKcs-expressing non-cancerous fibroblasts. Results obtained for NU7026 were confirmed by PRKDC knockdown in NGP cells. Taken together, the current study shows that DNA-PKcs is a promising target for neuroblastoma radiosensitization.
Collapse
Affiliation(s)
- M. Emmy M. Dolman
- Department of Oncogenomics, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- * E-mail:
| | - Ida van der Ploeg
- Department of Oncogenomics, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Jan Koster
- Department of Oncogenomics, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Laurel Tabe Bate-Eya
- Department of Oncogenomics, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Rogier Versteeg
- Department of Oncogenomics, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Huib N. Caron
- Department of Pediatric Oncology, Emma Kinderziekenhuis, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Jan J. Molenaar
- Department of Oncogenomics, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
14
|
Pal I, Dey KK, Chaurasia M, Parida S, Das S, Rajesh Y, Sharma K, Chowdhury T, Mandal M. Cooperative effect of BI-69A11 and celecoxib enhances radiosensitization by modulating DNA damage repair in colon carcinoma. Tumour Biol 2015; 37:6389-402. [DOI: 10.1007/s13277-015-4399-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 11/05/2015] [Indexed: 12/16/2022] Open
|
15
|
Phosphatidylinositol 3-kinase/Akt signaling as a key mediator of tumor cell responsiveness to radiation. Semin Cancer Biol 2015; 35:180-90. [PMID: 26192967 DOI: 10.1016/j.semcancer.2015.07.003] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 07/09/2015] [Accepted: 07/13/2015] [Indexed: 02/07/2023]
Abstract
The phosphatidylinositol 3-kinase (PI3K)/Akt pathway is a key cascade downstream of several protein kinases, especially membrane-bound receptor tyrosine kinases, including epidermal growth factor receptor (EGFR) family members. Hyperactivation of the PI3K/Akt pathway is correlated with tumor development, progression, poor prognosis, and resistance to cancer therapies, such as radiotherapy, in human solid tumors. Akt/PKB (Protein Kinase B) members are the major kinases that act downstream of PI3K, and these are involved in a variety of cellular functions, including growth, proliferation, glucose metabolism, invasion, metastasis, angiogenesis, and survival. Accumulating evidence indicates that activated Akt is one of the major predictive markers for solid tumor responsiveness to chemo/radiotherapy. DNA double-strand breaks (DNA-DSB), are the prime cause of cell death induced by ionizing radiation. Preclinical in vitro and in vivo studies have shown that constitutive activation of Akt and stress-induced activation of the PI3K/Akt pathway accelerate the repair of DNA-DSB and, consequently, lead to therapy resistance. Analyzing dysregulations of Akt, such as point mutations, gene amplification or overexpression, which results in the constitutive activation of Akt, might be of special importance in the context of radiotherapy outcomes. Such studies, as well as studies of the mechanism(s) by which activated Akt1 regulates repair of DNA-DSB, might help to identify combinations using the appropriate molecular targeting strategies with conventional radiotherapy to overcome radioresistance in solid tumors. In this review, we discuss the dysregulation of the components of upstream regulators of Akt as well as specific modifications of Akt isoforms that enhance Akt activity. Likewise, the mechanisms by which Akt interferes with repair of DNA after exposure to ionizing radiation, will be reviewed. Finally, the current status of Akt targeting in combination with radiotherapy will be discussed.
Collapse
|
16
|
Chen J. Signaling pathways in HPV-associated cancers and therapeutic implications. Rev Med Virol 2015; 25 Suppl 1:24-53. [PMID: 25752815 DOI: 10.1002/rmv.1823] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Revised: 10/15/2014] [Accepted: 12/27/2014] [Indexed: 12/19/2022]
Affiliation(s)
- Jiezhong Chen
- School of Biomedical Sciences and Australian Institute for Bioengineering and Nanotechnology; The University of Queensland; Brisbane Queensland Australia
| |
Collapse
|
17
|
Wong P, Houghton P, Kirsch DG, Finkelstein SE, Monjazeb AM, Xu-Welliver M, Dicker AP, Ahmed M, Vikram B, Teicher BA, Coleman CN, Machtay M, Curran WJ, Wang D. Combining targeted agents with modern radiotherapy in soft tissue sarcomas. J Natl Cancer Inst 2014; 106:dju329. [PMID: 25326640 DOI: 10.1093/jnci/dju329] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Improved understanding of soft-tissue sarcoma (STS) biology has led to better distinction and subtyping of these diseases with the hope of exploiting the molecular characteristics of each subtype to develop appropriately targeted treatment regimens. In the care of patients with extremity STS, adjunctive radiation therapy (RT) is used to facilitate limb and function, preserving surgeries while maintaining five-year local control above 85%. In contrast, for STS originating from nonextremity anatomical sites, the rate of local recurrence is much higher (five-year local control is approximately 50%) and a major cause of death and morbidity in these patients. Incorporating novel technological advancements to administer accurate RT in combination with novel radiosensitizing agents could potentially improve local control and overall survival. RT efficacy in STS can be increased by modulating biological pathways such as angiogenesis, cell cycle regulation, cell survival signaling, and cancer-host immune interactions. Previous experiences, advancements, ongoing research, and current clinical trials combining RT with agents modulating one or more of the above pathways are reviewed. The standard clinical management of patients with STS with pretreatment biopsy, neoadjuvant treatment, and primary surgery provides an opportune disease model for interrogating translational hypotheses. The purpose of this review is to outline a strategic vision for clinical translation of preclinical findings and to identify appropriate targeted agents to combine with radiotherapy in the treatment of STS from different sites and/or different histology subtypes.
Collapse
Affiliation(s)
- Philip Wong
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Peter Houghton
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - David G Kirsch
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Steven E Finkelstein
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Arta M Monjazeb
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Meng Xu-Welliver
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Adam P Dicker
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Mansoor Ahmed
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Bhadrasain Vikram
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Beverly A Teicher
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - C Norman Coleman
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Mitchell Machtay
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Walter J Curran
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW)
| | - Dian Wang
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montréal, Québec, Canada (PW); Research Institute at Nationwide Children's Hospital, Columbus, OH (PH); Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC (DGK); 21st Century Oncology Translational Research Consortium (TRC) Headquarters, Scottsdale, AZ (SEF); Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, CA (AMM); Department of Radiation Oncology, the Ohio State University, Columbus, OH (MXW); Department of Radiation Oncology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA (APD); Radiotherapy Development Branch & Molecular Radiation Therapeutics Branch, Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD (MA, CNC); Clinical Radiation Oncology Branch, National Cancer Institute, Bethesda, MD (BV); Molecular Pharmacology Branch, National Cancer Institute, Bethesda, MD (BAT); Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH (MM); Winship Cancer Institute, Woodruff Health Science Center, Emory University, Atlanta, GA (WJC); Department of Radiation Oncology, Rush University Medical Center, Chicago, IL (DW).
| |
Collapse
|
18
|
Wei L, Qu W, Sun J, Wang X, Lv L, Xie L, Song X. Knockdown of cancerous inhibitor of protein phosphatase 2A may sensitize NSCLC cells to cisplatin. Cancer Gene Ther 2014; 21:194-9. [PMID: 24874844 DOI: 10.1038/cgt.2014.18] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 04/09/2014] [Accepted: 04/09/2014] [Indexed: 11/09/2022]
Abstract
Cancerous inhibitor of protein phosphatase 2A (CIP2A) is a recently identified human oncoprotein that can stabilize some proteins by inhibiting degradation mediated by protein phosphatase 2A (PP2A) and it increases the proliferation of several cancer cells. Recent studies have highlighted a potential role for CIP2A in promoting tumor progression and metastasis. However, whether CIP2A could increase chemoresistance of cancer cells to chemotherapeutic agent cisplatin remains unclear. To determine whether CIP2A serves as a potential therapeutic target of human non-small-cell lung cancer (NSCLC), we utilized small interference RNA (siRNA) to knock down CIP2A expression in human NSCLC cells and analyzed their phenotypic changes. The data demonstrated that CIP2A silencing led to decreased proliferation, impaired clonogenicity and enhanced chemosensitivity and apoptosis to cisplatin in human NSCLC cells, as well as reduced Akt phosphorylation. In addition, overexpression of CIP2A diminished NSCLC cell chemosensitivity to cisplatin by inducing activation of Akt pathway, suggesting critical roles of CIP2A in NSCLC cell chemoresistance to cisplatin and rasing the possibility of CIP2A inhibition as a promising approach for lung cancer therapy.
Collapse
Affiliation(s)
- L Wei
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, China
| | - W Qu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, China
| | - J Sun
- Department of Pathology, Shandong Cancer Hospital and Institute, Jinan, China
| | - X Wang
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, China
| | - L Lv
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, China
| | - L Xie
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, China
| | - X Song
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, China
| |
Collapse
|
19
|
Palmieri M, Desai J, Sieber O. What is the future potential of the PI3K pathway in colorectal cancer treatment? COLORECTAL CANCER 2014. [DOI: 10.2217/crc.14.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Michelle Palmieri
- Systems Biology & Personalised Medicine Division, Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Faculty of Medicine, Dentistry & Health Sciences, Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Jayesh Desai
- Systems Biology & Personalised Medicine Division, Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Oncology, Royal Melbourne Hospital, Parkville & Western Hospital, Footscray, Victoria, Australia
| | - Oliver Sieber
- Systems Biology & Personalised Medicine Division, Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| |
Collapse
|
20
|
Lei Y, Li HX, Jin WS, Peng WR, Zhang CJ, Bu LJ, Du YY, Ma T, Sun GP. The radiosensitizing effect of Paeonol on lung adenocarcinoma by augmentation of radiation-induced apoptosis and inhibition of the PI3K/Akt pathway. Int J Radiat Biol 2013; 89:1079-86. [PMID: 23875954 DOI: 10.3109/09553002.2013.825058] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
PURPOSE To investigate the radiosensitizing effect and mechanism of action by the natural product Paeonol on lung adenocarcinoma both in vitro and in vivo. MATERIALS AND METHODS Two lung adenocarcinoma cell lines (human lung adenocarcinoma cell line A549 and mouse Lewis lung carcinoma (LLC) cell line) were chosen for this research. In order to select the experimental concentrations of Paeonol, cytotoxicity was determined using a MTT (3-[4,5-dimethylthiazol-2-yl]-2,5- diphenyltetrazolium bromide) assay. A clonogenic assay was performed to measure the radiosensitizing effects. Apoptosis was determined by the Tunel (terminal deoxynucleotidyl transferase-mediated dUTP nick and labeling) assay and flow cytometry. Protein expression was analyzed by Western blotting. To test the radiosensitizing effect in vivo, a transplanted tumor model was established. RESULTS The MTT assay showed that Paeonol inhibited proliferation of cells. Paeonol concentration ranged from an IC5 (5% inhibiting concentration) to an IC20 and was used at non-toxic concentrations for subsequent experiments. The clonogenic assay showed that Paeonol enhanced the radiosensitivity of cells. Data from the Tunel assay and flow cytometry verified that Paeonol enhanced radiation-induced apoptosis. Paeonol inhibited the activation of the PI3K/AKT (Phosphatidylinositol 3-kinase/ Protein Kinase B) pathway and down-regulated the expression of COX-2 (Cyclooxygenase-2) and Survivin. Paeonol (1718 mg/kg) combined with 10 Gy irradiation inhibited the growth of a transplanted tumor model in vivo, resulting in the longest tumor growth time, tumor growth delay and the highest inhibition ratio when compared with the radiotherapy alone group. CONCLUSIONS It is reported for the first time that Paeonol has a radiosensitizing effect on lung adenocarcinoma both in vitro and in vivo. This effect could be related to the augmentation of radiation-induced apoptosis and the inhibition of the PI3K/Akt signalling pathway and its downstream proteins: COX-2 and Survivin.
Collapse
Affiliation(s)
- Yu Lei
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University , Hefei
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
DNA hypermethylation biomarkers to predict response to cisplatin treatment, radiotherapy or chemoradiation: the present state of art. Cell Oncol (Dordr) 2012; 35:231-41. [DOI: 10.1007/s13402-012-0091-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2012] [Indexed: 12/20/2022] Open
|