1
|
Patil VM, Masand N, Verma S, Masand V. Chromones: Privileged scaffold in anticancer drug discovery. Chem Biol Drug Des 2021; 98:943-953. [PMID: 34519163 DOI: 10.1111/cbdd.13951] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/02/2021] [Accepted: 09/06/2021] [Indexed: 12/16/2022]
Abstract
In the design and discovery of anticancer drugs, various natural heterocyclic scaffolds have attracted considerable interest as privileged structures. For rational drug design, some of the natural scaffolds such as chromones have exhibited wide acceptability due to their drug-like properties. Among the approved anticancer drugs, the scaffolds with high selectivity for a small group of closely related targets are of importance. In the development of selective anticancer agents, the natural, as well as synthetic, can generate highly selective compounds toward cancer targets. The present manuscript includes more particularly the development of cancer inhibitors incorporating the chromone scaffold, with a strong emphasis on their molecular interactions in the anticancer mechanism. It also includes the structure-activity relationship studies and related examples of lead optimization.
Collapse
Affiliation(s)
- Vaishali M Patil
- Department of Pharmaceutical Chemistry, KIET School of Pharmacy, KIET Group of Institutions, Delhi-NCR, Ghaziabad, India
| | - Neeraj Masand
- Department of Pharmacy, Lala Lajpat Rai Memorial Medical College, Meerut, Uttar Pradesh, India
| | - Saroj Verma
- Department of Pharmaceutical Chemistry, SGT University, Gurugram, Haryana, India
| | - Vijay Masand
- Department of Chemistry, Vidya Bharati College, Amravati, Maharashtra, India
| |
Collapse
|
2
|
Su X, Liu J, Zhang H, Gu Q, Zhou X, Ji M, Yao D. Lenvatinib Promotes the Antitumor Effect of Doxorubicin in Anaplastic Thyroid Cancer. Onco Targets Ther 2020; 13:11183-11192. [PMID: 33173310 PMCID: PMC7646435 DOI: 10.2147/ott.s278349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 10/14/2020] [Indexed: 12/14/2022] Open
Abstract
Purpose Anaplastic thyroid cancer (ATC) is a kind of rare thyroid cancer with very poor prognosis. Doxorubicin has been approved in ATC treatment as a single agent, but monotherapy still shows no improvement of the total survival in advanced ATC. Lenvatinib was investigated with encouraging results in treating patients with radioiodine-refractory differentiated thyroid cancer (DTC). However, antitumor efficacy of combination therapy with lenvatinib and doxorubicin remains largely unclear. Materials and Methods The antitumor efficacy of combination therapy with lenvatinib and doxorubicin on ATC cell proliferation was assessed by the MTT assay and colony formation. Flow cytometry was employed to assess ATC cells’ apoptosis and cell cycle arrest in response to combination therapy. Transwell assay was used to test the migration and invasion in response to combination therapy. Xenograft models were used to test its in vivo antitumor activity. Results Lenvatinib monotherapy was less effective than doxorubicin in treating ATC cell lines and xenograft model. The combination therapy of lenvatinib and doxorubicin significantly inhibited ATC cell proliferation and tumor growth in nude mice, and induced cell apoptosis and cell cycle arrest as compared to lenvatinib or doxorubicin monotherapy. Conclusion Lenvatinib promotes the antitumor effect of doxorubicin in ATC cell and xenograft model. The lenvatinib/doxorubicin combination may be a potential candidate therapeutic approach for anaplastic thyroid cancer.
Collapse
Affiliation(s)
- Xi Su
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, People's Republic of China
| | - Jiaxin Liu
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, People's Republic of China
| | - Haihong Zhang
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, People's Republic of China
| | - Qingqing Gu
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, People's Republic of China
| | - Xinrui Zhou
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, People's Republic of China
| | - Meiju Ji
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, People's Republic of China
| | - Demao Yao
- Department of Geriatric Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, People's Republic of China
| |
Collapse
|
3
|
Xia X, Wang Q, Ye T, Liu Y, Liu D, Song S, Zheng C. NRF2/ABCB1-mediated efflux and PARP1-mediated dampening of DNA damage contribute to doxorubicin resistance in chronic hypoxic HepG2 cells. Fundam Clin Pharmacol 2019; 34:41-50. [PMID: 31420991 DOI: 10.1111/fcp.12505] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 06/09/2019] [Accepted: 08/14/2019] [Indexed: 12/30/2022]
Abstract
Transarterial chemoembolization (TACE)-induced hypoxia can trigger residual liver cancer cells to present a more aggressive phenotype associated with chemoresistance, but the underlying mechanisms are still unknown. In this study, the human liver cancer cell line HepG2 was pre-cultured in different oxygen environments to examine the possible mechanisms of hypoxia-induced doxorubicin resistance. Our study showed that HepG2 cells pre-cultured in a chronic intermittent hypoxic environment exhibited significant resistance to doxorubicin, evidenced by increased intracellular doxorubicin efflux, relatively higher cell proliferation, lower apoptosis, and decreased DNA damage. These changes were accompanied by high levels of NRF2 and ABCB1 under conditions of both chronic and acute hypoxia and PARP1 gene expression only under conditions of chronic hypoxia. SiRNA-mediated silencing of NRF2 gene expression downregulated the expression of ABCB1 and increased the intracellular doxorubicin accumulation and cell apoptosis both in acute and chronic hypoxic HepG2 cells. Moreover, silencing of PARP1 gene expression increased the doxorubicin-induced DNA damage and cell apoptosis in chronic hypoxic cells. On the basis of these findings, we concluded that NRF2/ABCB1-mediated efflux and PARP1-mediated DNA repair contribute to doxorubicin resistance in chronic hypoxic HepG2 cells.
Collapse
Affiliation(s)
- Xiangwen Xia
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Qi Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Tianhe Ye
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Yiming Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Dehan Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Songlin Song
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei, 430022, China
| |
Collapse
|
4
|
Tang JY, Xu YH, Lin LC, Ou-Yang F, Wu KH, Tsao LY, Yu TJ, Huang HW, Wang HR, Liu W, Chang HW. LY303511 displays antiproliferation potential against oral cancer cells in vitro and in vivo. ENVIRONMENTAL TOXICOLOGY 2019; 34:958-967. [PMID: 31115172 DOI: 10.1002/tox.22767] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 04/17/2019] [Accepted: 04/24/2019] [Indexed: 06/09/2023]
Abstract
LY303511 was developed as a negative control of LY294002 without pan-phosphoinositide 3-kinase (PI3K) inhibition. We hypothesize LY303511 generate reactive oxygen species (ROS) to induce apoptosis for killing oral cancer cells. In MTS assay, LY303511 dose-responsively decreases survival in three kinds of oral cancer cells but little damage to normal oral cells (HGF-1). Two oral cancer cells (CAL 27 and SCC-9) with highly sensitivity to LY303511 were used. In 7-aminoactinomycin D (7AAD) assay, LY303511 slightly increases subG1 population in oral cancer cells. In annexin V/7AAD and/or pancaspase assays, LY303511 induces apoptosis in oral cancer cells but HGF-1 cells remains in basal level. In oxidative stress, LY303511 induces ROS and mitochondrial superoxide in oral cancer cells. In 8-oxo-2'-deoxyguanosine assay, LY303511 induces oxidative DNA damage in oral cancer cells. In zebrafish model, LY303511 inhibits CAL 27-xenografted tumor growth. Therefore, LY303511 displays antiproliferation potential against oral cancer cells in vitro and in vivo.
Collapse
Affiliation(s)
- Jen-Yang Tang
- Department of Radiation Oncology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Yi-Hua Xu
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Li-Ching Lin
- Department of Radiation Oncology, Chi-Mei Foundation Medical Center, Tainan, Taiwan
- School of Medicine, Taipei Medical University, Taipei, Taiwan
- Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Fu Ou-Yang
- Division of Breast Surgery and Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Kuang-Han Wu
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Li-Yi Tsao
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tzu-Jung Yu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hurng-Wern Huang
- Institute of Biomedical Science, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Hui-Ru Wang
- Institute of Biomedical Science, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Wangta Liu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
5
|
Cui Y, Wang J, Liu S, Qu D, Jin H, Zhu L, Yang J, Zhang J, Li Q, Zhang Y, Yao Y. miR‐216a promotes breast cancer cell apoptosis by targeting
PKC
α. Fundam Clin Pharmacol 2019; 33:397-404. [PMID: 31119784 DOI: 10.1111/fcp.12481] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 04/22/2019] [Accepted: 05/10/2019] [Indexed: 12/28/2022]
Affiliation(s)
- Ying Cui
- Department of Radiation Oncology Harbin Medical University Cancer Hospital Heilongjiang 150081 China
| | - Jinghao Wang
- Department of Pharmacy the First Affiliated Hospital Jinan University Guangzhou 510630 China
| | - Shanshan Liu
- Department of Radiation Oncology Harbin Medical University Cancer Hospital Heilongjiang 150081 China
| | - Di Qu
- Department of Medical Oncology the Second Affiliated Hospital of Harbin Medical University Heilongjiang 150086 China
| | - Hong Jin
- Department of Gynecology Harbin Medical University Cancer Hospital Heilongjiang 150081 China
| | - Lin Zhu
- Department of Radiation Oncology Harbin Medical University Cancer Hospital Heilongjiang 150081 China
| | - Jiani Yang
- Department of Medical Oncology Harbin Medical University Cancer Hospital Heilongjiang 150081China
| | - Jingchun Zhang
- Department of Medical Oncology Harbin Medical University Cancer Hospital Heilongjiang 150081China
| | - Qingwei Li
- Department of Medical Oncology Harbin Medical University Cancer Hospital Heilongjiang 150081China
| | - Yanqiao Zhang
- Department of Medical Oncology Harbin Medical University Cancer Hospital Heilongjiang 150081China
| | - Yuanfei Yao
- Department of Medical Oncology Harbin Medical University Cancer Hospital Heilongjiang 150081China
| |
Collapse
|
6
|
Mues M, Karra L, Romero-Moya D, Wandler A, Hangauer MJ, Ksionda O, Thus Y, Lindenbergh M, Shannon K, McManus MT, Roose JP. High-Complexity shRNA Libraries and PI3 Kinase Inhibition in Cancer: High-Fidelity Synthetic Lethality Predictions. Cell Rep 2019; 27:631-647.e5. [PMID: 30970263 PMCID: PMC6690758 DOI: 10.1016/j.celrep.2019.03.045] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 10/11/2018] [Accepted: 03/13/2019] [Indexed: 12/24/2022] Open
Abstract
Deregulated signal transduction is a cancer hallmark, and its complexity and interconnectivity imply that combination therapy should be considered, but large data volumes that cover the complexity are required in user-friendly ways. Here, we present a searchable database resource of synthetic lethality with a PI3 kinase signal transduction inhibitor by performing a saturation screen with an ultra-complex shRNA library containing 30 independent shRNAs per gene target. We focus on Ras-PI3 kinase signaling with T cell leukemia as a screening platform for multiple clinical and experimental reasons. Our resource predicts multiple combination-based therapies with high fidelity, ten of which we confirmed with small molecule inhibitors. Included are biochemical assays, as well as the IPI145 (duvelisib) inhibitor. We uncover the mechanism of synergy between the PI3 kinase inhibitor GDC0941 (pictilisib) and the tubulin inhibitor vincristine and demonstrate broad synergy in 28 cell lines of 5 cancer types and efficacy in preclinical leukemia mouse trials.
Collapse
Affiliation(s)
- Marsilius Mues
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Laila Karra
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Damia Romero-Moya
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Anica Wandler
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Matthew J Hangauer
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Olga Ksionda
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yvonne Thus
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Marthe Lindenbergh
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kevin Shannon
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michael T McManus
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jeroen P Roose
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
7
|
Yang L, Ye F, Bao L, Zhou X, Wang Z, Hu P, Ouyang N, Li X, Shi Y, Chen G, Xia P, Chui M, Li W, Jia Y, Liu Y, Liu J, Ye J, Zhang Z, Bu H. Somatic alterations of TP53, ERBB2, PIK3CA and CCND1 are associated with chemosensitivity for breast cancers. Cancer Sci 2019; 110:1389-1400. [PMID: 30776175 PMCID: PMC6447848 DOI: 10.1111/cas.13976] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/24/2019] [Accepted: 02/08/2019] [Indexed: 02/05/2023] Open
Abstract
The correlation of genetic alterations with response to neoadjuvant chemotherapy (NAC) has not been fully revealed. In this study, we enrolled 247 breast cancer patients receiving anthracycline‐taxane‐based NAC treatment. A next generation sequencing (NGS) panel containing 36 hotspot breast cancer‐related genes was used in this study. Two different standards for the extent of pathologic complete response (pCR), ypT0/isypN0 and ypT0/is, were used as indicators for NAC treatment. TP53 mutation (n = 149, 60.3%), PIK3CA mutation (n = 109, 44.1%) and MYC amplification (n = 95, 38.5%) were frequently detected in enrolled cases. TP53 mutation (P = 0.019 for ypT0/isypN0 and P = 0.003 for ypT0/is) and ERBB2 amplification (P < 0.001 for both ypT0/isypN0 and ypT0/is) were related to higher pCR rates. PIK3CA mutation (P = 0.040 for ypT0/isypN0) and CCND2 amplification (P = 0.042 for ypT0/is) showed reduced sensitivity to NAC. Patients with MAPK pathway alteration had low pCR rates (P = 0.043 for ypT0/is). Patients with TP53 mutation (−) PIK3CA mutation (−) ERBB2 amplification (+) CCND1 amplification (−), TP53 mutation (+) PIK3CA mutation (−) ERBB2 amplification (+) CCND1 amplification (−) or TP53 mutation (+) PIK3CA mutation (+) ERBB2 amplification (+) CCND1 amplification (−)had significantly higher pCR rates (P < 0.05 for ypT0/isypN0 and ypT0/is) than wild type genotype tumors. Some cancer genetic alterations as well as pathway alterations were associated with chemosensitivity to NAC treatment. Our study may shed light on the molecular characteristics of breast cancer for prediction of NAC expectations when breast cancer is first diagnosed by biopsy.
Collapse
Affiliation(s)
- Libo Yang
- Department of Pathology, West China Hospital of Sichuan University, Chengdu, China.,Laboratory of Pathology, West China Hospital of Sichuan University, Chengdu, China
| | - Feng Ye
- Laboratory of Pathology, West China Hospital of Sichuan University, Chengdu, China
| | - Longlong Bao
- Department of Pathology, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Shanghai, China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Xiaoyan Zhou
- Department of Pathology, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Shanghai, China.,Institute of Pathology, Fudan University, Shanghai, China
| | - Zhe Wang
- Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Peizhen Hu
- Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Nengtai Ouyang
- Cellular & Molecular Diagnostics Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaojuan Li
- Cellular & Molecular Diagnostics Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yi Shi
- Department of Molecular Pathology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Gang Chen
- Department of Molecular Pathology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Peiyi Xia
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Pathology, School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Meiying Chui
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Pathology, School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Wencai Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Pathology, School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Ying Jia
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yueping Liu
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | | | - Junyi Ye
- Burning Rock Biotech, Guangzhou, China
| | - Zhe Zhang
- Burning Rock Biotech, Guangzhou, China
| | - Hong Bu
- Department of Pathology, West China Hospital of Sichuan University, Chengdu, China.,Laboratory of Pathology, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
8
|
Cai Q, Lin J, Zhang L, Lin S, Peng J. Chloroform fraction of Serratulae chinensis S. Moore suppresses proliferation and induces apoptosis via the phosphatidylinositide 3-kinase/Akt pathway in human gastric cancer cells. Oncol Lett 2018; 15:8871-8877. [PMID: 29928328 DOI: 10.3892/ol.2018.8366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 08/03/2017] [Indexed: 11/05/2022] Open
Abstract
The chloroform fraction of the folk Chinese medicine, Serratulae chinensis S. Moore (CSC) and its anti-inflammatory activity is well recognized. However, the molecular mechanisms underlying the beneficial anticancer effects of CSC remain largely unknown. The aim of the present study was to examine the effects of CSC on the regulation of cell proliferation and apoptosis in SGC-7901 gastric cancer cells, as well as to investigate the underlying molecular mechanisms involved. The results from the present study demonstrated that CSC treatment inhibited SGC-7901 cell viability and survival in a dose- and/or time-dependent manner. CSC treatment further induced the apoptosis of SGC-7901 cells, characterized by distinct chromatin condensation and fragmented nuclear morphology. In addition, CSC treatment suppressed protein kinase-B (Akt) phosphorylation and phosphatidylinositide 3-kinase (PI3K) expression in SGC-7901 cells, which in turn promoted cancer cell apoptosis and inhibited cell proliferation. Furthermore, CSC treatment altered the expression pattern of several key target genes of the PI3K/Akt signaling pathway through the downregulation of Cyclin D1, cyclin-dependent kinase-4 and B-cell lymphoma-2 and the upregulation of Bcl-2-associated X protein. Therefore, the results from the present study demonstrated that CSC suppressed cell survival and induced apoptosis in human gastric cancer cells, via targeting the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Qiaoyan Cai
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Jing Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Ling Zhang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Shan Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| |
Collapse
|
9
|
Niessner H, Kosnopfel C, Sinnberg T, Beck D, Krieg K, Wanke I, Lasithiotakis K, Bonin M, Garbe C, Meier F. Combined activity of temozolomide and the mTOR inhibitor temsirolimus in metastatic melanoma involves DKK1. Exp Dermatol 2018; 26:598-606. [PMID: 28423208 DOI: 10.1111/exd.13372] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2017] [Indexed: 02/03/2023]
Abstract
The BRAFV600E inhibitor vemurafenib achieves remarkable clinical responses in patients with BRAF-mutant melanoma, but its effects are limited by the onset of drug resistance. In the case of resistance, chemotherapy can still be applied as second line therapy. However, it yields low response rates and strategies are urgently needed to potentiate its effects. In a previous study, we showed that the inhibition of the PI3K-AKT-mTOR pathway significantly increases sensitivity of melanoma cells to chemotherapeutic drugs (J. Invest. Dermatol. 2009, 129, 1500). In this study, the combination of the mTOR inhibitor temsirolimus with the chemotherapeutic agent temozolomide significantly increases growth inhibition and apoptosis in melanoma cells compared to temsirolimus or temozolomide alone. The combination of temozolomide with temsirolimus is not only effective in established but also in newly isolated and vemurafenib-resistant metastatic melanoma cell lines. These effects are associated with the downregulation of the anti-apoptotic protein Mcl-1 and the upregulation of the Wnt antagonist Dickkopf homologue 1 (DKK1). Knock-down of DKK1 suppresses apoptosis induction by the combination of temsirolimus and temozolomide. These data suggest that the inhibition of the mTOR pathway increases sensitivity of melanoma cells towards temozolomide. Chemosensitisation is associated with enhanced expression of the Wnt antagonist DKK1.
Collapse
Affiliation(s)
- Heike Niessner
- Department of Dermatology, Division of Dermatooncology, University of Tübingen, Tübingen, Germany
| | - Corinna Kosnopfel
- Department of Dermatology, Division of Dermatooncology, University of Tübingen, Tübingen, Germany
| | - Tobias Sinnberg
- Department of Dermatology, Division of Dermatooncology, University of Tübingen, Tübingen, Germany
| | - Daniela Beck
- Department of Dermatology, Division of Dermatooncology, University of Tübingen, Tübingen, Germany
| | - Kathrin Krieg
- Department of Dermatology, Division of Dermatooncology, University of Tübingen, Tübingen, Germany
| | - Ines Wanke
- Department of Dermatology, Division of Dermatooncology, University of Tübingen, Tübingen, Germany
| | | | - Michael Bonin
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Claus Garbe
- Department of Dermatology, Division of Dermatooncology, University of Tübingen, Tübingen, Germany
| | - Friedegund Meier
- Department of Dermatology, Division of Dermatooncology, University of Tübingen, Tübingen, Germany.,Department of Dermatology, Carl Gustav Carus Medical Center, TU Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
| |
Collapse
|
10
|
Pillinger G, Loughran NV, Piddock RE, Shafat MS, Zaitseva L, Abdul-Aziz A, Lawes MJ, Bowles KM, Rushworth SA. Targeting PI3Kδ and PI3Kγ signalling disrupts human AML survival and bone marrow stromal cell mediated protection. Oncotarget 2018; 7:39784-39795. [PMID: 27174919 PMCID: PMC5129970 DOI: 10.18632/oncotarget.9289] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 04/16/2016] [Indexed: 02/06/2023] Open
Abstract
Phosphoinositide-3-kinase (PI3K) is an enzyme group, known to regulate key survival pathways in acute myeloid leukaemia (AML). It generates phosphatidylinositol-3,4,5-triphosphate, which provides a membrane docking site for protein kinaseB activation. PI3K catalytic p110 subunits are divided into 4 isoforms; α,β,δ and γ. The PI3Kδ isoform is always expressed in AML cells, whereas the frequency of PI3Kγ expression is highly variable. The functions of these individual catalytic enzymes have not been fully resolved in AML, therefore using the PI3K p110δ and p110γ-targeted inhibitor IPI-145 (duvelisib) and specific p110δ and p110γ shRNA, we analysed the role of these two p110 subunits in human AML blast survival. The results show that PI3Kδ and PI3Kγ inhibition with IPI-145 has anti-proliferative activity in primary AML cells by inhibiting the activity of AKT and MAPK. Pre-treatment of AML cells with IPI-145 inhibits both adhesion and migration of AML blasts to bone marrow stromal cells. Using shRNA targeted to the individual isoforms we demonstrated that p110δ-knockdown had a more significant anti-proliferative effect on AML cells, whereas targeting p110γ-knockdown significantly inhibited AML migration. The results demonstrate that targeting both PI3Kδ and PI3Kγ to inhibit AML-BMSC interactions provides a biologic rationale for the pre-clinical evaluation of IPI-145 in AML.
Collapse
Affiliation(s)
- Genevra Pillinger
- Department of Molecular Haematology, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Niamh V Loughran
- Department of Molecular Haematology, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Rachel E Piddock
- Department of Molecular Haematology, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Manar S Shafat
- Department of Molecular Haematology, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Lyubov Zaitseva
- Department of Molecular Haematology, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Amina Abdul-Aziz
- Department of Molecular Haematology, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Matthew J Lawes
- Department of Haematology, Norfolk and Norwich University Hospitals NHS Trust, Norwich, NR4 7UY, United Kingdom
| | - Kristian M Bowles
- Department of Molecular Haematology, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom.,Department of Haematology, Norfolk and Norwich University Hospitals NHS Trust, Norwich, NR4 7UY, United Kingdom
| | - Stuart A Rushworth
- Department of Molecular Haematology, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| |
Collapse
|
11
|
Zeng SX, Zhu Y, Ma AH, Yu W, Zhang H, Lin TY, Shi W, Tepper CG, Henderson PT, Airhart S, Guo JM, Xu CL, deVere White RW, Pan CX. The Phosphatidylinositol 3-Kinase Pathway as a Potential Therapeutic Target in Bladder Cancer. Clin Cancer Res 2017; 23:6580-6591. [PMID: 28808038 DOI: 10.1158/1078-0432.ccr-17-0033] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 07/07/2017] [Accepted: 08/08/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Activation of the PI3K pathway occurs in over 40% of bladder urothelial cancers. The aim of this study is to determine the therapeutic potential, the underlying action, and the resistance mechanisms of drugs targeting the PI3K pathway.Experimental Design: Urothelial cancer cell lines and patient-derived xenografts (PDXs) were analyzed for alterations of the PI3K pathway and for their sensitivity to the small-molecule inhibitor pictilisib alone and in combination with cisplatin and/or gemcitabine. Potential predictive biomarkers for pictilisib were evaluated, and RNA sequencing was performed to explore drug resistance mechanisms.Results: The bladder cancer cell line TCCSUP, which harbors a PIK3CA E545K mutation, was sensitive to pictilisib compared to cell lines with wild-type PIK3CA Pictilisib exhibited stronger antitumor activity in bladder cancer PDX models with PI3KCA H1047R mutation or amplification than the control PDX model. Pictilisib synergized with cisplatin and/or gemcitabine in vitro, significantly delayed tumor growth, and prolonged survival compared with single-drug treatment in the PDX models. The phosphorylation of ribosomal protein S6 correlated with response to pictilisib both in vitro and in vivo, and could potentially serve as a biomarker to predict response to pictilisib. Pictilisib activated the compensatory MEK/ERK pathway that likely contributed to pictilisib resistance, which was reversed by cotreatment with the RAF inhibitor sorafenib. RNA sequencing of tumors resistant to treatment suggested that LSP1 downregulation correlated with drug resistance.Conclusions: These preclinical results provide new insights into the therapeutic potential of targeting the PI3K pathway for the treatment of bladder cancer. Clin Cancer Res; 23(21); 6580-91. ©2017 AACR.
Collapse
Affiliation(s)
- Shu-Xiong Zeng
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, University of California Davis, Sacramento, California.,Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yanjun Zhu
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, University of California Davis, Sacramento, California.,Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ai-Hong Ma
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, California
| | - Weimin Yu
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, University of California Davis, Sacramento, California.,Department of Urology, Renmin Hospital, Wuhan University, Wuhan, Hubei Province, China
| | - Hongyong Zhang
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, University of California Davis, Sacramento, California
| | - Tzu-Yin Lin
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, University of California Davis, Sacramento, California
| | - Wei Shi
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, University of California Davis, Sacramento, California.,Department of Urology, Xijing Hospital, The Fourth Military Medical University, Shanxi Province, China
| | - Clifford G Tepper
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, California
| | - Paul T Henderson
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, University of California Davis, Sacramento, California
| | | | - Jian-Ming Guo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chuan-Liang Xu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China.
| | - Ralph W deVere White
- Department of Urology, University of California Davis Comprehensive Cancer Center, Sacramento, California
| | - Chong-Xian Pan
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, University of California Davis, Sacramento, California. .,Department of Urology, University of California Davis Comprehensive Cancer Center, Sacramento, California.,VA Northern California Health Care System, Mather, California
| |
Collapse
|
12
|
Reis J, Gaspar A, Milhazes N, Borges F. Chromone as a Privileged Scaffold in Drug Discovery: Recent Advances. J Med Chem 2017; 60:7941-7957. [PMID: 28537720 DOI: 10.1021/acs.jmedchem.6b01720] [Citation(s) in RCA: 229] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The use of privileged structures in drug discovery has proven to be an effective strategy, allowing the generation of innovative hits/leads and successful optimization processes. Chromone is recognized as a privileged structure and a useful template for the design of novel compounds with potential pharmacological interest, particularly in the field of neurodegenerative, inflammatory, and infectious diseases as well as diabetes and cancer. This perspective provides the reader with an update of an earlier article entitled "Chromone: A Valid Scaffold in Medicinal Chemistry" ( Chem. Rev. 2014 , 114 , 4960 - 4992 ) and is mainly focused on chromones of biological interest, including those isolated from natural sources. Moreover, as drug repurposing is becoming an attractive drug discovery approach, recent repurposing studies of chromone-based drugs are also reported.
Collapse
Affiliation(s)
- Joana Reis
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto , Porto 4169-007, Portugal
| | - Alexandra Gaspar
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto , Porto 4169-007, Portugal
| | - Nuno Milhazes
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto , Porto 4169-007, Portugal
| | - Fernanda Borges
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto , Porto 4169-007, Portugal
| |
Collapse
|
13
|
|
14
|
Aboubakr EM, Taye A, Aly OM, Gamal-Eldeen AM, El-Moselhy MA. Enhanced anticancer effect of Combretastatin A-4 phosphate when combined with vincristine in the treatment of hepatocellular carcinoma. Biomed Pharmacother 2017; 89:36-46. [PMID: 28214686 DOI: 10.1016/j.biopha.2017.02.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 02/02/2017] [Accepted: 02/07/2017] [Indexed: 01/16/2023] Open
Abstract
Tubulin targeting agents have received considerable interest as a potential tumor-selective vascular disrupting agents, which represent another avenue for cancer growing therapeutic opportunities. Hence, the present study was conducted to investigate the anti-tumor activity of Combretastatin A-4 phosphate (CA4-P) and vincristine against hepatocellular carcinoma in rats, by individual administration and in combination. In vitro study was conducted using human hepatocellular carcinoma cell lines, showed that CA4-P and vincristine have a potent cell cytotoxic and tubulin inhibitory effect. In addition, a remarkable synergistic effect was observed by the simultaneous application of both drugs. Whereas in vivo study was conducted using model of rat liver cancer initiated with DENA and promoted by CCl4, showed that CA4-P and vincristine were significantly decreased liver relative weight, number of hepatic nodules and there relative volumes, tubulin content of the hepatic tissue, GSH and AFP. On the other hand, co-administration of both drugs exhibited significant further decrements in these parameters. Whereas a marked increase in MDA, carbonyl content and TNF-α inside hepatic tissue were observed in the treated groups and these increments were more prominent by co-administration of both drugs. In conclusion CA4-P showed a potential anti-cancer activity against hepatocellular carcinoma and this effect was greatly enhanced by co-administration with vincristine. Additionally, our new findings provided an important evidence that the anticancer activity of drugs with a narrow therapeutic window such as vincristine can be greatly improved by its co-administration with CA4-P providing more enhanced activity with less side effects.
Collapse
Affiliation(s)
- Esam M Aboubakr
- Department of Pharmacology and Toxicology, South Valley University, 6 Km, Qena Safaga Road, Qena, Egypt.
| | - Ashraf Taye
- Department of Pharmacology and Toxicology, Minia University, Ard Shalaby, Misr-Aswan Road, El-Minia, Egypt.
| | - Omar M Aly
- Department of Medicinal Chemistry, Minia University, Ard Shalaby, Misr-Aswan Road, El-Minia, Egypt.
| | - Amira M Gamal-Eldeen
- Cancer Biology, Center of excellence for advanced sciences, National Research Center, El Buhouth St., Dokki, Cairo, Egypt.
| | - Mohamed A El-Moselhy
- Department of Pharmacology and Toxicology, Minia University, Ard Shalaby, Misr-Aswan Road, El-Minia, Egypt.
| |
Collapse
|
15
|
Liu A, Wu J, Li A, Bi W, Liu T, Cao L, Liu Y, Dong L. The inhibitory mechanism of Cordyceps sinensis on cigarette smoke extract-induced senescence in human bronchial epithelial cells. Int J Chron Obstruct Pulmon Dis 2016; 11:1721-31. [PMID: 27555762 PMCID: PMC4968689 DOI: 10.2147/copd.s107396] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVES Cellular senescence is a state of irreversible growth arrest induced either by telomere shortening (replicative senescence) or stress. The bronchial epithelial cell is often injured by inhaled toxic substances, such as cigarette smoke. In the present study, we investigated whether exposure to cigarette smoke extract (CSE) induces senescence of bronchial epithelial cells; and Cordyceps sinensis mechanism of inhibition of CSE-induced cellular senescence. METHODS Human bronchial epithelial cells (16HBE cells) cultured in vitro were treated with CSE and/or C. sinensis. p16, p21, and senescence-associated-galactosidase activity were used to detect cellular senescence with immunofluorescence, quantitative polymerase chain reaction, and Western blotting. Reactive oxygen species (ROS), PI3K/AKT/mTOR and their phosphorylated proteins were examined to testify the activation of signaling pathway by ROS fluorescent staining and Western blotting. Then, inhibitors of ROS and PI3K were used to further confirm the function of this pathway. RESULTS Cellular senescence was upregulated by CSE treatment, and C. sinensis can decrease CSE-induced cellular senescence. Activation of ROS/PI3K/AKT/mTOR signaling pathway was enhanced by CSE treatment, and decreased when C. sinensis was added. Blocking ROS/PI3K/AKT/mTOR signaling pathway can attenuate CSE-induced cellular senescence. CONCLUSION CSE can induce cellular senescence in human bronchial epithelial cells, and ROS/PI3K/AKT/mTOR signaling pathway may play an important role in this process. C. sinensis can inhibit the CSE-induced senescence.
Collapse
Affiliation(s)
- Ailing Liu
- Department of Pulmonary Diseases, Qilu Hospital, Shandong University, Jinan, Shandong, People’s Republic of China
- Department of Pulmonary Diseases, Weihai Municipal Hospital, Weihai, Shandong, People’s Republic of China
| | - Jinxiang Wu
- Department of Pulmonary Diseases, Qilu Hospital, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Aijun Li
- Department of Pulmonary Diseases, Weihai Municipal Hospital, Weihai, Shandong, People’s Republic of China
| | - Wenxiang Bi
- Institute of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Tian Liu
- Department of Pulmonary Diseases, Qilu Hospital, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Liuzhao Cao
- Department of Pulmonary Diseases, Qilu Hospital, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Yahui Liu
- Department of Pulmonary Diseases, Qilu Hospital, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Liang Dong
- Department of Pulmonary Diseases, Qilu Hospital, Shandong University, Jinan, Shandong, People’s Republic of China
| |
Collapse
|
16
|
Ung MH, Varn FS, Cheng C. IDEA: Integrated Drug Expression Analysis-Integration of Gene Expression and Clinical Data for the Identification of Therapeutic Candidates. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2015; 4:415-25. [PMID: 26312165 PMCID: PMC4544055 DOI: 10.1002/psp4.51] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 04/30/2015] [Indexed: 12/14/2022]
Abstract
Cancer drug discovery is an involved process spanning efforts from several fields of study and typically requires years of research and development. However, the advent of high-throughput genomic technologies has allowed for the use of in silico, genomics-based methods to screen drug libraries and accelerate drug discovery. Here we present a novel approach to computationally identify drug candidates for the treatment of breast cancer. In particular, we developed a Drug Regulatory Score similarity metric to evaluate gene expression profile similarity, in the context of drug treatment, and incorporated time-to-event patient survival information to develop an integrated analysis pipeline: Integrated Drug Expression Analysis (IDEA). We were able to predict drug candidates that have been known and those that have not been known in the literature to exhibit anticancer effects. Overall, our method enables quick preclinical screening of drug candidates for breast cancer and other diseases by using the most important indicator of drug efficacy: survival.
Collapse
Affiliation(s)
- M H Ung
- Department of Genetics, Geisel School of Medicine at Dartmouth Hanover, New Hampshire, USA
| | - F S Varn
- Department of Genetics, Geisel School of Medicine at Dartmouth Hanover, New Hampshire, USA
| | - C Cheng
- Department of Genetics, Geisel School of Medicine at Dartmouth Hanover, New Hampshire, USA ; Institute for Quantitative Biomedical Sciences, Geisel School of Medicine at Dartmouth Lebanon, New Hampshire, USA ; Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth Lebanon, New Hampshire, USA
| |
Collapse
|
17
|
Ethiraj P, Veerappan K, Samuel S, Sivapatham S. Inhibitory effects of interferon-β on hepatocellular carcinoma HepG2 via Akt/STAT phosphorylation. Fundam Clin Pharmacol 2015; 29:278-285. [DOI: 10.1111/fcp.12115] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Purushoth Ethiraj
- Department of Medical Research; SRM Medical College Hospital and Research Centre; SRM University; Kattankulathur 603203 Tamilnadu India
| | - Karpagam Veerappan
- Department of Biochemistry; VRR Institute of Biomedical Science; Chennai 600056 Tamilnadu India
| | - Shila Samuel
- Department of Biochemistry; VRR Institute of Biomedical Science; Chennai 600056 Tamilnadu India
| | - Sundaresan Sivapatham
- Department of Medical Research; SRM Medical College Hospital and Research Centre; SRM University; Kattankulathur 603203 Tamilnadu India
| |
Collapse
|
18
|
Cioce M, Canino C, Goparaju C, Yang H, Carbone M, Pass HI. Autocrine CSF-1R signaling drives mesothelioma chemoresistance via AKT activation. Cell Death Dis 2014; 5:e1167. [PMID: 24722292 PMCID: PMC5424113 DOI: 10.1038/cddis.2014.136] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 02/21/2014] [Accepted: 02/28/2014] [Indexed: 01/02/2023]
Abstract
Clinical management of malignant pleural mesothelioma (MPM) is very challenging because of the uncommon resistance of this tumor to chemotherapy. We report here increased expression of macrophage colony-stimulating-factor-1-receptor (M-CSF/CSF-1R) mRNA in mesothelioma versus normal tissue specimens and demonstrate that CSF-1R expression identifies chemoresistant cells of mesothelial nature in both primary cultures and mesothelioma cell lines. By using RNAi or ligand trapping, we demonstrate that the chemoresistance properties of those cells depend on autocrine CSF-1R signaling. At the single-cell level, the isolated CSF-1Rpos cells exhibit a complex repertoire of pluripotency, epithelial–mesenchymal transition and detoxifying factors, which define a clonogenic, chemoresistant, precursor-like cell sub-population. The simple activation of CSF-1R in untransformed mesothelial cells is sufficient to confer clonogenicity and resistance to pemetrexed, hallmarks of mesothelioma. In addition, this induced a gene expression profile highly mimicking that observed in the MPM cells endogenously expressing the receptor and the ligands, suggesting that CSF-1R expression is mainly responsible for the phenotype of the identified cell sub-populations. The survival of CSF1Rpos cells requires active AKT (v-akt murine thymoma viral oncogene homolog 1) signaling, which contributed to increased levels of nuclear, transcriptionally competent β-catenin. Inhibition of AKT reduced the transcriptional activity of β-catenin-dependent reporters and sensitized the cells to senescence-induced clonogenic death after pemetrexed treatment. This work expands what is known on the non-macrophage functions of CSF-1R and its role in solid tumors, and suggests that CSF-1R signaling may have a critical pathogenic role in a prototypical, inflammation-related cancer such as MPM and therefore may represent a promising target for therapeutic intervention.
Collapse
Affiliation(s)
- M Cioce
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Langone Medical Center, New York University, New York, NY, USA
| | - C Canino
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Langone Medical Center, New York University, New York, NY, USA
| | - C Goparaju
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Langone Medical Center, New York University, New York, NY, USA
| | - H Yang
- University of Hawaii Cancer Center, John A Burns School of Medicine, University of Hawaii, Honolulu, HA, USA
| | - M Carbone
- University of Hawaii Cancer Center, John A Burns School of Medicine, University of Hawaii, Honolulu, HA, USA
| | - H I Pass
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Langone Medical Center, New York University, New York, NY, USA
| |
Collapse
|