1
|
Zhao S, Liu Y, Wang H, Wang J, Zhang J, Liu Y, Ma D. Mechanisms and progress of LncRNAs in prostate cancer development and diagnostic therapy. Int Urol Nephrol 2025:10.1007/s11255-025-04497-z. [PMID: 40266504 DOI: 10.1007/s11255-025-04497-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/28/2025] [Indexed: 04/24/2025]
Abstract
Prostate cancer (PCa) is a leading cause of cancer-related morbidity and mortality in men worldwide. Despite advancements in diagnosis and treatment, challenges such as late-stage detection, therapeutic resistance, and the complexity of castration-resistant prostate cancer (CRPC) persist. Long non-coding RNAs (LncRNAs) play critical roles in PCa progression through epigenetic regulation, transcriptional and post-transcriptional modulation, and immune response regulation. This review highlights the molecular mechanisms by which LncRNAs influence PCa development, treatment resistance, and immune regulation, emphasizing their potential as biomarkers and therapeutic targets. We also discuss future research directions to advance precision medicine in PCa.
Collapse
Affiliation(s)
- Shihan Zhao
- School of Basic Medical College, Beihua University, Jilin, 132013, China
| | - Yuqi Liu
- School of Basic Medical College, Beihua University, Jilin, 132013, China
| | - Han Wang
- School of Basic Medical College, Beihua University, Jilin, 132013, China
| | - Jiayi Wang
- School of Basic Medical College, Beihua University, Jilin, 132013, China
| | - Jihong Zhang
- The Pathology Department of Affiliated Hospital, Beihua University, Jilin, 132013, China
| | - Yanbo Liu
- School of Basic Medical College, Beihua University, Jilin, 132013, China.
| | - Dongrui Ma
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| |
Collapse
|
2
|
Zhang Q, Peng J, Zhang Y, Liu J, He D, Zhao Y, Wang X, Li C, Kong Y, Wang R, Mao F, Wang C, Wang Q, Zhang M, Wang J, Yang HS, Liu X. The kinase PLK1 promotes Hedgehog signaling-dependent resistance to the antiandrogen enzalutamide in metastatic prostate cancer. Sci Signal 2025; 18:eadi5174. [PMID: 40100956 PMCID: PMC11988940 DOI: 10.1126/scisignal.adi5174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/20/2024] [Accepted: 02/18/2025] [Indexed: 03/20/2025]
Abstract
Enzalutamide, a second-generation androgen receptor inhibitor (also known as an antiandrogen), is used to treat patients with metastatic castration-resistant prostate cancer (CRPC). Tumors often acquire resistance to enzalutamide. Tumor progression and enzalutamide resistance are associated with decreased abundance of the tumor suppressor PDCD4. In normal dividing cells, PDCD4 abundance is low when that of the kinase PLK1 is high. In this study, we found that PLK1 acted on PDCD4 to promote enzalutamide resistance in CRPC cells in culture and in mice via a mechanism that revealed an effective combination therapy. PLK1 phosphorylated PDCD4 at Ser239, leading to its degradation and consequently inducing the transcriptional activation of Hedgehog (Hh) signaling by c-MYC. Hh signaling supports tumor cell proliferation and stemness by inducing the enzyme UDP-glucuronosyltransferase 2B15 (UGT2B15), which promotes the metabolic clearance of drugs and steroid hormones. Thus, this pathway may circumvent androgen receptor dependence, thereby reducing cellular sensitivity to enzalutamide. Knocking down UGT2B15 enhanced enzalutamide-induced cell apoptosis and growth arrest in a PDCD4-dependent manner. Combining enzalutamide with the clinically approved Hh pathway inhibitor vismodegib inhibited cell growth and promoted apoptosis in enzalutamide-resistant cell cultures and xenografts in vivo. Our findings reveal a mechanism of PLK1-mediated enzalutamide resistance and suggest a potential therapeutic strategy to overcome this resistance in prostate cancer.
Collapse
Affiliation(s)
- Qiongsi Zhang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Jia Peng
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Yanquan Zhang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Jinghui Liu
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Daheng He
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Yue Zhao
- Gilbert S. Omenn Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xinyi Wang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Chaohao Li
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Yifan Kong
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Ruixin Wang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Fengyi Mao
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Chi Wang
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Qing Wang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Min Zhang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Jianlin Wang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Hsin-Sheng Yang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Xiaoqi Liu
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
3
|
Li Y, Tang X, Wang B, Chen M, Zheng J, Chang K. Current landscape of exosomal non-coding RNAs in prostate cancer: Modulators and biomarkers. Noncoding RNA Res 2024; 9:1351-1362. [PMID: 39247145 PMCID: PMC11380467 DOI: 10.1016/j.ncrna.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/12/2024] [Accepted: 07/18/2024] [Indexed: 09/10/2024] Open
Abstract
Prostate cancer (PCa) has the highest frequency of diagnosis among solid tumors and ranks second as the primary cause of cancer-related deaths. Non-coding RNAs (ncRNAs), such as microRNAs, long non-coding RNAs and circular RNAs, frequently exhibit dysregulation and substantially impact the biological behavior of PCa. Compared with circulating ncRNAs, ncRNAs loaded into exosomes are more stable because of protection by the lipid bilayer. Furthermore, exosomal ncRNAs facilitate the intercellular transfer of molecules and information. Increasing evidence suggests that exosomal ncRNAs hold promising potential in the progression, diagnosis and prognosis of PCa. This review aims to discuss the functions of exosomal ncRNAs in PCa, evaluate their possible applications as clinical biomarkers and therapeutic targets, and provide a comprehensive overview of the ncRNAs regulatory network in PCa. We also identified ncRNAs that can be utilized as biomarkers for diagnosis, staging, grading and prognosis assessment in PCa. This review offers researchers a fresh perspective on the functions of exosomal ncRNAs in PCa and provides additional options for its diagnosis, progression monitoring, and prognostic prediction.
Collapse
Affiliation(s)
- Yongxing Li
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, PR China
- School of Medicine, Chongqing University, Chongqing, 400030, PR China
| | - Xiaoqi Tang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| | - Binpan Wang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| | - Ming Chen
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, PR China
- School of Medicine, Chongqing University, Chongqing, 400030, PR China
- State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| | - Kai Chang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
- State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| |
Collapse
|
4
|
Li Q, Wang Y, Chen J, Zeng K, Wang C, Guo X, Hu Z, Hu J, Liu B, Xiao J, Zhou P. Machine learning based androgen receptor regulatory gene-related random forest survival model for precise treatment decision in prostate cancer. Heliyon 2024; 10:e37256. [PMID: 39296076 PMCID: PMC11407950 DOI: 10.1016/j.heliyon.2024.e37256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 09/21/2024] Open
Abstract
Background It has been demonstrated that aberrant androgen receptor (AR) signaling contributes to the pathogenesis of prostate cancer (PCa). To date, the most efficacious strategy for the treatment of PCa remains to target the AR signaling axis. However, numerous PCa patients still face the issue of overtreatment or undertreatment. The establishment of a precise risk prediction model is urgently needed to distinguish patients with high-risk and select appropriate treatment modalities. Methods In this study, a consensus AR regulatory gene-related signature (ARS) was developed by integrating a total of 101 algorithm combinations of 10 machine learning algorithms. We evaluated the value of ARS in predicting patient prognosis and the therapeutic effects of the various treatments. Additionally, we conducted a screening of therapeutic targets and agents for high-risk patients, followed by the verification in vitro and in vivo. Results ARS was an independent risk factor for biochemical recurrence and distant metastasis in PCa patients. The enhanced and consistent prognostic predictive capability of ARS across various platforms was confirmed when compared with 44 previously published signatures. More importantly, PCa patients in the ARShigh group benefit more from PARP inhibitors and immunotherapy, while chemotherapy, radiotherapy, and AR-targeted therapy are more effective for ARSlow patients. The results of in silico screening suggest that AURKB could potentially serve as a promising therapeutic target for ARShigh patients. Conclusions Collectively, this prediction model based on AR regulatory genes holds great clinical translational potential to solve the dilemma of treatment choice and identify potential novel therapeutic targets in PCa.
Collapse
Affiliation(s)
- Qinyu Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Yanan Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Junjie Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Kai Zeng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Chengwei Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Xiangdong Guo
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Zhiquan Hu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Jia Hu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Bo Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Jun Xiao
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Peng Zhou
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
5
|
Jang A, Lanka SM, Ruan HT, Kumar HLS, Jia AY, Garcia JA, Mian OY, Barata PC. Novel therapies for metastatic prostate cancer. Expert Rev Anticancer Ther 2023; 23:1251-1263. [PMID: 38030394 DOI: 10.1080/14737140.2023.2290197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/28/2023] [Indexed: 12/01/2023]
Abstract
INTRODUCTION Patients with metastatic prostate cancer, especially in the castrate-resistant setting, have a poor prognosis. Many agents have been approved for metastatic prostate cancer, such as androgen receptor pathway inhibitors, taxane-based chemotherapy, radiopharmaceuticals, and immunotherapy. However, prostate cancer remains the leading cause of cancer deaths in nonsmoking men. Fortunately, many more novel agents are under investigation. AREAS COVERED We provide an overview of the broad group of novel therapies for metastatic prostate cancer, with an emphasis on active and recruiting clinical trials that have been recently published and/or presented at national or international meetings. EXPERT OPINION The future for patients with metastatic prostate cancer is promising, with further development of novel therapies such as radiopharmaceuticals. Based on a growing understanding of prostate cancer biology, novel agents are being designed to overcome resistance to approved therapies. There are many trials using novel agents either as monotherapy or in combination with already approved agents with potential to further improve outcomes for men with advanced prostate cancer.
Collapse
Affiliation(s)
- Albert Jang
- Division of Solid Tumor Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Sree M Lanka
- Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Hui Ting Ruan
- Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Hamsa L S Kumar
- Division of Solid Tumor Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Angela Y Jia
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Jorge A Garcia
- Division of Solid Tumor Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Omar Y Mian
- Translational Hematology and Oncology Research, Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Pedro C Barata
- Division of Solid Tumor Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Case Comprehensive Cancer Center, Cleveland, OH, USA
| |
Collapse
|
6
|
Chuang HW, Pan JH, Cai YX, Rupa D, Huang TS, Kuo TC, Lin CW, Chen CW, Lin CC, Lee HS, Yuan TC. Reciprocal regulation of CIP2A and AR expression in prostate cancer cells. Discov Oncol 2022; 13:87. [PMID: 36098827 PMCID: PMC9470804 DOI: 10.1007/s12672-022-00552-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 08/30/2022] [Indexed: 11/17/2022] Open
Abstract
Cancerous inhibitor of protein phosphatase 2A (CIP2A) is an oncoprotein overexpressed in human malignancies, including prostate cancer (PCa). In this study, we aimed to explore the oncogenic function of CIP2A in PCa cells and its underlying mechanism. We showed that 63.3% (38/60 cases) of PCa tissues exhibited a high CIP2A immunostaining, compared to 25% (3/12 cases) of BPH samples (p = 0.023). Furthermore, the protein level of CIP2A was positively correlated with patients' short survival time and nuclear AR levels in PCa tissues. Compared to PZ-HPV-7, an immortalized prostate cell line, androgen-sensitive LNCaP C-33, androgen-independent LNCaP C-81, or 22Rv1 cells exhibited a high CIP2A level, associated with high protein and phosphorylation levels of AR. While AR expression and activity modulated CIP2A expression, manipulating CIP2A expression in PCa cells regulated their AR protein levels and proliferation. The reduction of CIP2A expression also enhanced the sensitivity of PCa cells toward Enzalutamide treatment. Our data further showed that depletion of polo-kinase 1 (PLK1) expression or activity in C-81 or 22Rv1 cells caused reduced protein levels of c-Myc and AR. Notably, inhibition of PLK1 activity could abolish CIP2A-promoted expressions in c-Myc, AR, and prostate-specific antigen (PSA) in C-33 cells under an androgen-deprived condition, suggesting the role of PLK1 activity in CIP2A-promoted AR expression. In summary, our data showed the existence of a novel regulation between CIP2A and AR protein levels, which is critical for promoting PCa malignancy. Thus, CIP2A could serve as a therapeutic target for PCa.
Collapse
Affiliation(s)
- Hao-Wen Chuang
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, No. 386, Dazhong 1st Rd, Zuoying Dist, Kaohsiung, 813414, Taiwan, ROC
| | - Jian-Hua Pan
- Department of Life Science, National Dong Hwa University, No. 1, Sec. 2, Da Hsueh Rd., Shoufeng, Hualien, 974301, Taiwan, ROC
| | - Yi-Xuan Cai
- Department of Life Science, National Dong Hwa University, No. 1, Sec. 2, Da Hsueh Rd., Shoufeng, Hualien, 974301, Taiwan, ROC
| | - Darius Rupa
- Department of Life Science, National Dong Hwa University, No. 1, Sec. 2, Da Hsueh Rd., Shoufeng, Hualien, 974301, Taiwan, ROC
| | - Ting-Syuan Huang
- Department of Life Science, National Dong Hwa University, No. 1, Sec. 2, Da Hsueh Rd., Shoufeng, Hualien, 974301, Taiwan, ROC
| | - Tzu-Chien Kuo
- Department of Life Science, National Dong Hwa University, No. 1, Sec. 2, Da Hsueh Rd., Shoufeng, Hualien, 974301, Taiwan, ROC
| | - Chiao-Wen Lin
- Department of Life Science, National Dong Hwa University, No. 1, Sec. 2, Da Hsueh Rd., Shoufeng, Hualien, 974301, Taiwan, ROC
| | - Chi-Wei Chen
- Department of Life Science, National Dong Hwa University, No. 1, Sec. 2, Da Hsueh Rd., Shoufeng, Hualien, 974301, Taiwan, ROC
| | - Chia-Chin Lin
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, No. 386, Dazhong 1st Rd, Zuoying Dist, Kaohsiung, 813414, Taiwan, ROC
| | - Herng-Sheng Lee
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, No. 386, Dazhong 1st Rd, Zuoying Dist, Kaohsiung, 813414, Taiwan, ROC
| | - Ta-Chun Yuan
- Department of Life Science, National Dong Hwa University, No. 1, Sec. 2, Da Hsueh Rd., Shoufeng, Hualien, 974301, Taiwan, ROC.
| |
Collapse
|
7
|
Mao F, Kong Y, Liu J, Rao X, Li C, Donahue K, Zhang Y, Jones K, Zhang Q, Xu W, Liu X. Diptoindonesin G antagonizes AR signaling and enhances the efficacy of antiandrogen therapy in prostate cancer. Prostate 2022; 82:917-932. [PMID: 35322879 PMCID: PMC9035130 DOI: 10.1002/pros.24336] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 11/22/2021] [Accepted: 12/14/2021] [Indexed: 01/27/2023]
Abstract
BACKGROUND The androgen receptor (AR) signaling pathway has been well demonstrated to play a crucial role in the development, progression, and drug resistance of prostate cancer. Although the current anti-androgen therapy could significantly benefit prostate cancer patients initially, the efficacy of the single drug usually lasts for a relatively short period, as drug resistance quickly emerges. METHODS We have performed an unbiased bioinformatics analysis using the RNA-seq results in 22Rv1 cells to identify the cell response toward Dip G treatment. The RNA-seq results were validated by qRT-PCR. Protein levels were detected by western blot or staining. Cell viability was measured by Aquabluer and colony formation assay. RESULTS Here, we identified that Diptoindonesin G (Dip G), a natural extracted compound, could promote the proteasome degradation of AR and polo-like kinase 1 (PLK1) through modulating the activation of CHIP E3 ligase. Administration of Dip G has shown a profound efficiency in the suppression of AR and PLK1, not only in androgen-dependent LNCaP cells but also in castration-resistant and enzalutamide-resistant cells in a CHIP-dependent manner. Through co-targeting the AR signaling, Dip G robustly improved the efficacy of HSP90 inhibitors and enzalutamide in both human prostate cancer cells and in vivo xenograft mouse model. CONCLUSIONS Our results revealed that Dip G-mediated AR degradation would be a promising and valuable therapeutic strategy in the clinic.
Collapse
Affiliation(s)
- Fengyi Mao
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40506, USA
| | - Yifan Kong
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40506, USA
| | - Jinghui Liu
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40506, USA
| | - Xiongjian Rao
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40506, USA
| | - Chaohao Li
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40506, USA
| | - Kristine Donahue
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Yanquan Zhang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40506, USA
| | - Katelyn Jones
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40506, USA
| | - Qiongsi Zhang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40506, USA
| | - Wei Xu
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Xiaoqi Liu
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40506, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY 40506, USA
- To whom correspondence should be addressed: Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40506, USA. Tel: (859) 562-2006;
| |
Collapse
|
8
|
Cunningham CE, MacAuley MJ, Vizeacoumar FS, Abuhussein O, Freywald A, Vizeacoumar FJ. The CINs of Polo-Like Kinase 1 in Cancer. Cancers (Basel) 2020; 12:cancers12102953. [PMID: 33066048 PMCID: PMC7599805 DOI: 10.3390/cancers12102953] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Many alterations specific to cancer cells have been investigated as targets for targeted therapies. Chromosomal instability is a characteristic of nearly all cancers that can limit response to targeted therapies by ensuring the tumor population is not genetically homogenous. Polo-like Kinase 1 (PLK1) is often up regulated in cancers and it regulates chromosomal instability extensively. PLK1 has been the subject of much pre-clinical and clinical studies, but thus far, PLK1 inhibitors have not shown significant improvement in cancer patients. We discuss the numerous roles and interactions of PLK1 in regulating chromosomal instability, and how these may provide an avenue for identifying targets for targeted therapies. As selective inhibitors of PLK1 showed limited clinical success, we also highlight how genetic interactions of PLK1 may be exploited to tackle these challenges. Abstract Polo-like kinase 1 (PLK1) is overexpressed near ubiquitously across all cancer types and dysregulation of this enzyme is closely tied to increased chromosomal instability and tumor heterogeneity. PLK1 is a mitotic kinase with a critical role in maintaining chromosomal integrity through its function in processes ranging from the mitotic checkpoint, centrosome biogenesis, bipolar spindle formation, chromosome segregation, DNA replication licensing, DNA damage repair, and cytokinesis. The relation between dysregulated PLK1 and chromosomal instability (CIN) makes it an attractive target for cancer therapy. However, clinical trials with PLK1 inhibitors as cancer drugs have generally displayed poor responses or adverse side-effects. This is in part because targeting CIN regulators, including PLK1, can elevate CIN to lethal levels in normal cells, affecting normal physiology. Nevertheless, aiming at related genetic interactions, such as synthetic dosage lethal (SDL) interactions of PLK1 instead of PLK1 itself, can help to avoid the detrimental side effects associated with increased levels of CIN. Since PLK1 overexpression contributes to tumor heterogeneity, targeting SDL interactions may also provide an effective strategy to suppressing this malignant phenotype in a personalized fashion.
Collapse
Affiliation(s)
- Chelsea E. Cunningham
- Department of Pathology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (M.J.M.); (F.S.V.)
- Correspondence: (C.E.C.); (A.F.); (F.J.V.); Tel.: +1-(306)-327-7864 (C.E.C.); +1-(306)-966-5248 (A.F.); +1-(306)-966-7010 (F.J.V.)
| | - Mackenzie J. MacAuley
- Department of Pathology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (M.J.M.); (F.S.V.)
| | - Frederick S. Vizeacoumar
- Department of Pathology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (M.J.M.); (F.S.V.)
| | - Omar Abuhussein
- College of Pharmacy, University of Saskatchewan, 104 Clinic Place, Saskatoon, SK S7N 2Z4, Canada;
| | - Andrew Freywald
- Department of Pathology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (M.J.M.); (F.S.V.)
- Correspondence: (C.E.C.); (A.F.); (F.J.V.); Tel.: +1-(306)-327-7864 (C.E.C.); +1-(306)-966-5248 (A.F.); +1-(306)-966-7010 (F.J.V.)
| | - Franco J. Vizeacoumar
- Department of Pathology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; (M.J.M.); (F.S.V.)
- College of Pharmacy, University of Saskatchewan, 104 Clinic Place, Saskatoon, SK S7N 2Z4, Canada;
- Cancer Research, Saskatchewan Cancer Agency, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada
- Correspondence: (C.E.C.); (A.F.); (F.J.V.); Tel.: +1-(306)-327-7864 (C.E.C.); +1-(306)-966-5248 (A.F.); +1-(306)-966-7010 (F.J.V.)
| |
Collapse
|
9
|
Guan X, Sun D, Lu E, Urrutia JA, Reiter RE, Rettig M, Evans CP, Lara P, Gleave M, Beer TM, Thomas GV, Huang J, Aggarwal RR, Quigley DA, Foye A, Chen WS, Youngren J, Weinstein AS, Stuart JM, Feng FY, Small EJ, Xia Z, Alumkal JJ. Copy Number Loss of 17q22 Is Associated with Enzalutamide Resistance and Poor Prognosis in Metastatic Castration-Resistant Prostate Cancer. Clin Cancer Res 2020; 26:4616-4624. [PMID: 32727885 PMCID: PMC7484240 DOI: 10.1158/1078-0432.ccr-19-2303] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 05/24/2020] [Accepted: 06/29/2020] [Indexed: 12/22/2022]
Abstract
PURPOSE The purpose of this study was to measure genomic changes that emerge with enzalutamide treatment using analyses of whole-genome sequencing and RNA sequencing. EXPERIMENTAL DESIGN One hundred and one tumors from men with metastatic castration-resistant prostate cancer (mCRPC) who had not been treated with enzalutamide (n = 64) or who had enzalutamide-resistant mCRPC (n = 37) underwent whole genome sequencing. Ninety-nine of these tumors also underwent RNA sequencing. We analyzed the genomes and transcriptomes of these mCRPC tumors. RESULTS Copy number loss was more common than gain in enzalutamide-resistant tumors. Specially, we identified 124 protein-coding genes that were more commonly lost in enzalutamide-resistant samples. These 124 genes included eight putative tumor suppressors located at nine distinct genomic regions. We demonstrated that focal deletion of the 17q22 locus that includes RNF43 and SRSF1 was not present in any patient with enzalutamide-naïve mCRPC but was present in 16% (6/37) of patients with enzalutamide-resistant mCRPC. 17q22 loss was associated with lower RNF43 and SRSF1 expression and poor overall survival from time of biopsy [median overall survival of 19.3 months in 17q22 intact vs. 8.9 months in 17q22 loss, HR, 3.44 95% confidence interval (CI), 1.338-8.867, log-rank P = 0.006]. Finally, 17q22 loss was linked with activation of several targetable factors, including CDK1/2, Akt, and PLK1, demonstrating the potential therapeutic relevance of 17q22 loss in mCRPC. CONCLUSIONS Copy number loss is common in enzalutamide-resistant tumors. Focal deletion of chromosome 17q22 defines a previously unappreciated molecular subset of enzalutamide-resistant mCRPC associated with poor clinical outcome.
Collapse
Affiliation(s)
- Xiangnan Guan
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Duanchen Sun
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Eric Lu
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Joshua A Urrutia
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Robert Evan Reiter
- Institute of Urologic Oncology, University of California Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Center, Department of Urology, University of California Los Angeles, Los Angeles, California
| | - Matthew Rettig
- Jonsson Comprehensive Cancer Center, Department of Urology, University of California Los Angeles, Los Angeles, California
- VA Greater Los Angeles, Department of Medicine, Los Angeles, California
| | - Christopher P Evans
- University of California Davis Comprehensive Cancer Center, Sacramento, California
| | - Primo Lara
- University of California Davis Comprehensive Cancer Center, Sacramento, California
| | - Martin Gleave
- Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tomasz M Beer
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - George V Thomas
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Jiaoti Huang
- Duke University School of Medicine, Durham, North Carolina
| | - Rahul R Aggarwal
- University of California San Francisco, San Francisco, California
| | - David A Quigley
- University of California San Francisco, San Francisco, California
| | - Adam Foye
- University of California San Francisco, San Francisco, California
| | - William S Chen
- University of California San Francisco, San Francisco, California
| | - Jack Youngren
- University of California San Francisco, San Francisco, California
| | | | | | - Felix Y Feng
- University of California San Francisco, San Francisco, California
| | - Eric J Small
- University of California San Francisco, San Francisco, California
| | - Zheng Xia
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon.
| | - Joshi J Alumkal
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon.
- University of Michigan Rogel Cancer Center, Ann Arbor, Michigan
| |
Collapse
|
10
|
Hsu SK, Chang WT, Lin IL, Chen YF, Padalwar NB, Cheng KC, Teng YN, Wang CH, Chiu CC. The Role of Necroptosis in ROS-Mediated Cancer Therapies and Its Promising Applications. Cancers (Basel) 2020; 12:E2185. [PMID: 32764483 PMCID: PMC7465132 DOI: 10.3390/cancers12082185] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 07/30/2020] [Indexed: 02/07/2023] Open
Abstract
Over the past decades, promising therapies targeting different signaling pathways have emerged. Among these pathways, apoptosis has been well investigated and targeted to design diverse chemotherapies. However, some patients are chemoresistant to these therapies due to compromised apoptotic cell death. Hence, exploring alternative treatments aimed at different mechanisms of cell death seems to be a potential strategy for bypassing impaired apoptotic cell death. Emerging evidence has shown that necroptosis, a caspase-independent form of cell death with features between apoptosis and necrosis, can overcome the predicament of drug resistance. Furthermore, previous studies have also indicated that there is a close correlation between necroptosis and reactive oxygen species (ROS); both necroptosis and ROS play significant roles both under human physiological conditions such as the regulation of inflammation and in cancer biology. Several small molecules used in experiments and clinical practice eliminate cancer cells via the modulation of ROS and necroptosis. The molecular mechanisms of these promising therapies are discussed in detail in this review.
Collapse
Affiliation(s)
- Sheng-Kai Hsu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Wen-Tsan Chang
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - I-Ling Lin
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Yih-Fung Chen
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Nitin Balkrushna Padalwar
- Department of Chemistry, National Institute of Technology Tiruchirappalli, Tiruchirappalli 620015, Tamilnadu, India;
| | - Kai-Chun Cheng
- Department of Ophthalmology, Kaohsiung Municipal Hsiaokang Hospital, Kaohsiung 812, Taiwan;
- Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Yen-Ni Teng
- Department of Biological Sciences and Technology, National University of Tainan, Tainan 700, Taiwan;
| | - Chi-Huei Wang
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- The Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
11
|
Hu J, Deng J, Cao R, Xiong S, Guo J. LncRNA GAS5 participates in the regulation of dexamethasone on androgen receptor -negative and -positive prostate cancer cell proliferation. Mol Cell Probes 2020; 53:101607. [PMID: 32470500 DOI: 10.1016/j.mcp.2020.101607] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/15/2020] [Accepted: 05/25/2020] [Indexed: 11/17/2022]
Abstract
BACKGROUND Androgen receptor (AR) and long non-coding RNAs (lncRNA) play important roles in the initiation and progression of prostate cancer (PCa). The present study was designed to investigate whether lncRNA growth arrest-specific 5 (GAS5) is involved in the regulation of dexamethasone on the proliferation of AR+ PCa and AR- PCa cell lines. METHODS Cell proliferation and cell cycle distribution were assessed using MTT assay and flow cytometry, respectively. GAS5 expression was examined by quantitative real-time PCR. AR protein level was examined by Western blot. RNA immunoprecipitation and RNA pull-down were performed to analyze the binding of GAS5 to AR. RESULTS In AR- PCa cell line PC3, dexamethasone upregulated GAS5 expression, induced cell cycle arrest in the G0/G1 phase and inhibited cell proliferation, which were enhanced by GAS5 overexpression and attenuated by GAS5 silencing. However, in AR+ PCa cell line 22Rv1, dexamethasone had no obvious effects on GAS5 expression, cell cycle distribution and cell proliferation. AR was localized in the cytoplasm and bound to GAS5, counteracting the proliferation-inhibitory effect of GAS5. CONCLUSION Taken together, GAS5 participates in the regulation of dexamethasone on the proliferation of AR+ PCa and AR- PCa cell lines.
Collapse
Affiliation(s)
- Jieping Hu
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Jun Deng
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Runfu Cao
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Shida Xiong
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Ju Guo
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
12
|
Shao C, Yu B, Liu Y. Androgen receptor splicing variant 7: Beyond being a constitutively active variant. Life Sci 2019; 234:116768. [PMID: 31445027 DOI: 10.1016/j.lfs.2019.116768] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/12/2019] [Accepted: 08/13/2019] [Indexed: 01/01/2023]
Abstract
In prostate cancer development, the androgen receptor (AR) signaling plays a crucial role during both formation of early prostate lesions and progression to the lethal, incurable castration resistant stage. Accordingly, numerous approaches have been developed to inhibit AR activity including androgen deprivation therapy, application of the AR antagonists as well as the use of taxanes. However, these treatments, although effective initially, resistance inevitably occur for most of the patients within several years and limiting the therapeutic efficacy. Of note, alterations and reactivation of the AR signaling pathway have been demonstrated as the major reasons for the observed resistance. Accumulating evidences have suggested that synthesis of AR splicing variants, in particular, the constitutively active AR-V7, is one of the most important mechanisms that contribute to the abnormal AR signaling. In addition, clinical data also highlight the potential of using AR-V7 as a predictive biomarker and a therapeutic target in metastatic castration resistant prostate cancer (mCRPC). In this review, we summarize the recent findings concerning the specific role of AR-V7 in CRPC progression, drug resistance and its potential value in clinical assessment.
Collapse
Affiliation(s)
- Chen Shao
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Bingbing Yu
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yanan Liu
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China.
| |
Collapse
|
13
|
Shin SB, Woo SU, Yim H. Cotargeting Plk1 and androgen receptor enhances the therapeutic sensitivity of paclitaxel-resistant prostate cancer. Ther Adv Med Oncol 2019; 11:1758835919846375. [PMID: 31156720 PMCID: PMC6515847 DOI: 10.1177/1758835919846375] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 04/04/2019] [Indexed: 12/21/2022] Open
Abstract
Backgrounds: Despite the clinical success of taxanes, they still have limitations, such as chemoresistance. To overcome the limitations of paclitaxel, genetic alterations and targeting effects of altered genes were observed in paclitaxel-resistant cancer. Because paclitaxel-resistant cancer shows high levels of Plk1, a promising target in chemotherapy, the effectiveness of Plk1 inhibitors in paclitaxel-resistant cancer cells has been investigated. Methods: Paclitaxel-resistant cancer cells were developed by exposure of stepwise escalating levels of paclitaxel. Genetic alterations were detected by quantitative reverse transcription polymerase chain reaction (qRT-PCR) and immunoblotting. Using a cell viability assay, combined targeting effects for Plk1 and androgen receptor (AR) were determined. Clinical data were analyzed to understand the relationship between Plk1 and AR in prostate cancer patients. Results: Treatment with Plk1 inhibitors markedly reduced the expression of MDR1, MRP1, and Plk1 in the paclitaxel-resistant cancer. Among Plk1 inhibitors, genistein, recently found as a direct Plk1 inhibitor, tended to be more effective in the paclitaxel-resistant prostate cancer than the parental cancer cells, which was related to the suppression of the AR, as well as inhibition of Plk1 activity. A combination of Plk1 inhibitors and AR antagonist bicalutamide exhibited a synergistic effect in LNCaPTXR, as well as LNCaP cells, by inhibiting Plk1 and AR. Analysis of clinical data provides evidence for the relevance between Plk1 and AR in prostate cancer patients, showing that Plk1 and AR are strong predictors of poor survival rates. Conclusions: We suggest that cotargeting Plk1 and AR would be effective in advanced chemoresistant prostate cancer cells to overcome the limitations associated with paclitaxel.
Collapse
Affiliation(s)
- Sol-Bi Shin
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do, Korea
| | - Sang-Uk Woo
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do, Korea
| | - Hyungshin Yim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do 15588, Korea
| |
Collapse
|
14
|
Sundaresan L, Kumar P, Chatterjee S. Mechanistic insights into the differential effects of thalidomide and lenalidomide in metastatic prostate cancer. Future Oncol 2018; 14:2383-2401. [DOI: 10.2217/fon-2018-0090] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Aim: To understand why thalidomide and lenalidomide exhibit different responses in metastatic prostate cancer (mPCa) treatment. Methods: We analyzed the perturbation signatures of thalidomide, lenalidomide, flutamide treated mPCa cell line from Library of Integrated Network-based Cellular Signatures database and transcriptome of docetaxel-treated mPCa patients. Results: Flutamide and docetaxel downregulated ‘Steroid Biosynthesis’, ‘Cell cycle’ and PCa specific transcription factor networks. Thalidomide inhibited ‘Cell cycle’ and ‘E2F network’, possibly accounting for its synergistic effects with docetaxel. Conversely, lenalidomide promoted ‘Cell cycle’ and ‘Cholesterol biosynthesis’. Conclusion: Hence, we propose that lenalidomide upregulates cholesterol synthesis followed by enhanced rate of cell cycle, thereby nurturing a hyperproliferative tumor microenvironment. In summary, this study offers a possible explanation for the differential outcomes in the treatment of mPCa with thalidomide and lenalidomide.
Collapse
Affiliation(s)
- Lakshmikirupa Sundaresan
- AU-KBC Research Center, Anna University, Chennai, India
- Department of Biotechnology, Anna University, Chennai, India
| | - Pavitra Kumar
- AU-KBC Research Center, Anna University, Chennai, India
| | - Suvro Chatterjee
- AU-KBC Research Center, Anna University, Chennai, India
- Department of Biotechnology, Anna University, Chennai, India
| |
Collapse
|
15
|
Fucic A, Aghajanyan A, Culig Z, Le Novere N. Systems Oncology: Bridging Pancreatic and Castrate Resistant Prostate Cancer. Pathol Oncol Res 2018; 25:1269-1277. [PMID: 30220022 DOI: 10.1007/s12253-018-0467-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 09/03/2018] [Indexed: 12/31/2022]
Abstract
Large investments by pharmaceutical companies in the development of new antineoplastic drugs have not been resulting in adequate advances of new therapies. Despite the introduction of new methods, technologies, translational medicine and bioinformatics, the usage of collected knowledge is unsatisfactory. In this paper, using examples of pancreatic ductal adenocarcinoma (PaC) and castrate-resistant prostate cancer (CRPC), we proposed a concept showing that, in order to improve applicability of current knowledge in oncology, the re-clustering of clinical and scientific data is crucial. Such an approach, based on systems oncology, would include bridging of data on biomarkers and pathways between different cancer types. Proposed concept would introduce a new matrix, which enables combining of already approved therapies between cancer types. Paper provides a (a) detailed analysis of similarities in mechanisms of etiology and progression between PaC and CRPC, (b) diabetes as common hallmark of both cancer types and (c) knowledge gaps and directions of future investigations. Proposed horizontal and vertical matrix in cancer profiling has potency to improve current antineoplastic therapy efficacy. Systems biology map using Systems Biology Graphical Notation Language is used for summarizing complex interactions and similarities of mechanisms in biology of PaC and CRPC.
Collapse
Affiliation(s)
- A Fucic
- Institute for Medical Research and Occupational Health, Ksaverska c 2, 10000, Zagreb, Croatia.
| | - A Aghajanyan
- Institute of Medicine, Peoples' Friendship University of Russia, Moscow, Russian Federation
| | - Z Culig
- Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | | |
Collapse
|
16
|
Mao F, Li J, Luo Q, Wang R, Kong Y, Carlock C, Liu Z, Elzey BD, Liu X. Plk1 Inhibition Enhances the Efficacy of BET Epigenetic Reader Blockade in Castration-Resistant Prostate Cancer. Mol Cancer Ther 2018; 17:1554-1565. [PMID: 29716963 PMCID: PMC6030429 DOI: 10.1158/1535-7163.mct-17-0945] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 02/07/2018] [Accepted: 04/25/2018] [Indexed: 01/30/2023]
Abstract
Polo-like kinase 1 (Plk1), a crucial regulator of cell-cycle progression, is overexpressed in multiple types of cancers and has been proven to be a potent and promising target for cancer treatment. In case of prostate cancer, we once showed that antineoplastic activity of Plk1 inhibitor is largely due to inhibition of androgen receptor (AR) signaling. However, we also discovered that Plk1 inhibition causes activation of the β-catenin pathway and increased expression of c-MYC, eventually resulting in resistance to Plk1 inhibition. JQ1, a selective small-molecule inhibitor targeting the amino-terminal bromodomains of BRD4, has been shown to dramatically inhibit c-MYC expression and AR signaling, exhibiting antiproliferative effects in a range of cancers. Because c-MYC and AR signaling are essential for prostate cancer initiation and progression, we aim to test whether targeting Plk1 and BRD4 at the same time is an effective approach to treat prostate cancer. Herein, we show that a combination of Plk1 inhibitor GSK461364A and BRD4 inhibitor JQ1 had a strong synergistic effect on castration-resistant prostate cancer (CRPC) cell lines, as well as in CRPC xenograft tumors. Mechanistically, the synergistic effect is likely due to two reasons: (i) Plk1 inhibition results in the accumulation of β-catenin in the nucleus, thus elevation of c-MYC expression, whereas JQ1 treatment directly suppresses c-MYC transcription; (ii) Plk1 and BRD4 dual inhibition acts synergistically in inhibition of AR signaling. Mol Cancer Ther; 17(7); 1554-65. ©2018 AACR.
Collapse
Affiliation(s)
- Fengyi Mao
- Department of Biochemistry, Purdue University, West Lafayette, Indiana
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana
| | - Jie Li
- Department of Biochemistry, Purdue University, West Lafayette, Indiana
| | - Qian Luo
- Department of Biochemistry, Purdue University, West Lafayette, Indiana
| | - Ruixin Wang
- Department of Biochemistry, Purdue University, West Lafayette, Indiana
| | - Yifan Kong
- Department of Biochemistry, Purdue University, West Lafayette, Indiana
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana
| | - Colin Carlock
- Department of Biochemistry, Purdue University, West Lafayette, Indiana
| | - Zian Liu
- Department of Biochemistry, Purdue University, West Lafayette, Indiana
| | - Bennet D Elzey
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana
| | - Xiaoqi Liu
- Department of Biochemistry, Purdue University, West Lafayette, Indiana.
- Center for Cancer Research, Purdue University, West Lafayette, Indiana
| |
Collapse
|
17
|
Rondeau G, Abedinpour P, Chrastina A, Pelayo J, Borgstrom P, Welsh J. Differential gene expression induced by anti-cancer agent plumbagin is mediated by androgen receptor in prostate cancer cells. Sci Rep 2018; 8:2694. [PMID: 29426892 PMCID: PMC5807367 DOI: 10.1038/s41598-018-20451-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/15/2018] [Indexed: 12/24/2022] Open
Abstract
Treatment of mice harboring PTEN-P2 tumors in the prostate or on prostate tissue in vivo with 5-hydroxy-2-methyl-1,4-naphthoquinone, also known as plumbagin, results in tumor regression in castrated mice, but not in intact mice. This suggested that dihydrotestosterone (DHT) production in the testes may prevent cell death due to plumbagin treatment, but the underlying mechanism is not understood. We performed RNA-seq analysis on cells treated with combinations of plumbagin and DHT, and analyzed differential gene expression, to gain insight into the interactions between androgen and plumbgin. DHT and plumbagin synergize to alter the expression of many genes that are not differentially regulated by either single agent when used alone. These experiments revealed that, for many genes, increases in mRNAs caused by DHT are sharply down-regulated by plumbagin, and that many transcripts change in response to plumbagin in a DHT-dependent manner. This suggests that androgen receptor mediates some of the effects of plumbagin on gene expression.
Collapse
Affiliation(s)
- Gaelle Rondeau
- Vaccine Research Institute of San Diego, 3030 Bunker Hill Street, Suite 200, San Diego, CA, 92109, USA
| | - Parisa Abedinpour
- Vaccine Research Institute of San Diego, 3030 Bunker Hill Street, Suite 200, San Diego, CA, 92109, USA
| | - Adrian Chrastina
- Vaccine Research Institute of San Diego, 3030 Bunker Hill Street, Suite 200, San Diego, CA, 92109, USA
| | - Jennifer Pelayo
- Vaccine Research Institute of San Diego, 3030 Bunker Hill Street, Suite 200, San Diego, CA, 92109, USA
| | - Per Borgstrom
- Vaccine Research Institute of San Diego, 3030 Bunker Hill Street, Suite 200, San Diego, CA, 92109, USA
- Pellficure Pharmaceuticals, Inc., 2325 Camino del Collado, La Jolla, CA, 92037, USA
| | - John Welsh
- Vaccine Research Institute of San Diego, 3030 Bunker Hill Street, Suite 200, San Diego, CA, 92109, USA.
| |
Collapse
|
18
|
Weiss GJ, Jameson G, Von Hoff DD, Valsasina B, Davite C, Di Giulio C, Fiorentini F, Alzani R, Carpinelli P, Di Sanzo A, Galvani A, Isacchi A, Ramanathan RK. Phase I dose escalation study of NMS-1286937, an orally available Polo-Like Kinase 1 inhibitor, in patients with advanced or metastatic solid tumors. Invest New Drugs 2018; 36:85-95. [PMID: 28726132 DOI: 10.1007/s10637-017-0491-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 07/07/2017] [Indexed: 12/21/2022]
Abstract
Background Pharmacological inhibition of polo-like kinase 1 (PLK1) represents a new approach for the treatment of solid tumors. This study was aimed at determining the first cycle dose-limiting toxicities (DLTs) and related maximum tolerated dose (MTD) of NMS-1286937, a selective ATP-competitive PLK1-specific inhibitor. Secondary objectives included evaluation of its safety and pharmacokinetic (PK) profile in plasma, its antitumor activity, and its ability to modulate intracellular targets in biopsied tissue. Methods This was a Phase I, open-label, dose-escalation trial in patients with advanced/metastatic solid tumors. A treatment cycle comprised 5 days of oral administration followed by 16 days of rest, for a total of 21 days (3-week cycle). Results Nineteen of 21 enrolled patients with confirmed metastatic disease received study medication. No DLTs occurred at the first 3 dose levels (6, 12, and 24 mg/m2/day). At the subsequent dose level (48 mg/m2/day), 2 of 3 patients developed DLTs. An intermediate level of 36 mg/m2/day was therefore investigated. Four patients were treated and two DLTs were observed. After further cohort expansion, the MTD and recommended phase II dose (RP2D) were determined to be 24 mg/m2/day. Disease stabilization, observed in several patients, was the best treatment response observed. Hematological toxicity (mostly thrombocytopenia and neutropenia) was the major DLT. Systemic exposure to NMS-1286937 increased with dose and was comparable between two cycles of treatment following oral administration of the drug. Conclusions This study successfully identified the MTD and DLTs for NMS-1286937 and characterized its safety profile.
Collapse
Affiliation(s)
- Glen J Weiss
- Western Regional Medical Center, Cancer Treatment Centers of America, 14200 W Celebrate Life Way, Goodyear, AZ, 85338, USA.
- Virginia G. Piper Cancer Centers at Scottsdale Healthcare, Scottsdale, AZ, USA.
| | - Gayle Jameson
- Virginia G. Piper Cancer Centers at Scottsdale Healthcare, Scottsdale, AZ, USA
| | - Daniel D Von Hoff
- Virginia G. Piper Cancer Centers at Scottsdale Healthcare, Scottsdale, AZ, USA
| | | | - Cristina Davite
- CLInical Organization for Strategies and Solutions (CLIOSS) S.r.l, Mayo Scottsdale, Nerviano, Italy
| | - Claudia Di Giulio
- CLInical Organization for Strategies and Solutions (CLIOSS) S.r.l, Mayo Scottsdale, Nerviano, Italy
| | | | | | | | - Alessandro Di Sanzo
- CLInical Organization for Strategies and Solutions (CLIOSS) S.r.l, Mayo Scottsdale, Nerviano, Italy
| | | | | | - Ramesh K Ramanathan
- Virginia G. Piper Cancer Centers at Scottsdale Healthcare, Scottsdale, AZ, USA
| |
Collapse
|
19
|
Gu P, Chen X, Xie R, Han J, Xie W, Wang B, Dong W, Chen C, Yang M, Jiang J, Chen Z, Huang J, Lin T. lncRNA HOXD-AS1 Regulates Proliferation and Chemo-Resistance of Castration-Resistant Prostate Cancer via Recruiting WDR5. Mol Ther 2017; 25:1959-1973. [PMID: 28487115 DOI: 10.1016/j.ymthe.2017.04.016] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 04/07/2017] [Accepted: 04/12/2017] [Indexed: 02/01/2023] Open
Abstract
Castration-resistant prostate cancer (CRPC) that occurs after the failure of androgen deprivation therapy is the leading cause of deaths in prostate cancer patients. Thus, there is an obvious and urgent need to fully understand the mechanism of CRPC and discover novel therapeutic targets. Long noncoding RNAs (lncRNAs) are crucial regulators in many human cancers, yet their potential roles and molecular mechanisms in CRPC are poorly understood. In this study, we discovered that an lncRNA HOXD-AS1 is highly expressed in CRPC cells and correlated closely with Gleason score, T stage, lymph nodes metastasis, and progression-free survival. Knockdown of HOXD-AS1 inhibited the proliferation and chemo-resistance of CRPC cells in vitro and in vivo. Furthermore, we identified several cell cycle, chemo-resistance, and castration-resistance-related genes, including PLK1, AURKA, CDC25C, FOXM1, and UBE2C, that were activated transcriptionally by HOXD-AS1. Further investigation revealed that HOXD-AS1 recruited WDR5 to directly regulate the expression of target genes by mediating histone H3 lysine 4 tri-methylation (H3K4me3). In conclusion, our findings indicate that HOXD-AS1 promotes proliferation, castration resistance, and chemo-resistance in prostate cancer by recruiting WDR5. This sheds a new insight into the regulation of CRPC by lncRNA and provides a potential approach for the treatment of CRPC.
Collapse
Affiliation(s)
- Peng Gu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Xu Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Ruihui Xie
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jinli Han
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Weibin Xie
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Bo Wang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Wen Dong
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Changhao Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Meihua Yang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Junyi Jiang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Department of Clinical Laboratory, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Ziyue Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Department of Pediatric Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jian Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| | - Tianxin Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| |
Collapse
|
20
|
Bengoechea-Alonso MT, Ericsson J. The phosphorylation-dependent regulation of nuclear SREBP1 during mitosis links lipid metabolism and cell growth. Cell Cycle 2016; 15:2753-65. [PMID: 27579997 PMCID: PMC5053579 DOI: 10.1080/15384101.2016.1220456] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 07/13/2016] [Accepted: 07/31/2016] [Indexed: 01/02/2023] Open
Abstract
The SREBP transcription factors are major regulators of lipid metabolism. Disturbances in lipid metabolism are at the core of several health issues facing modern society, including cardiovascular disease, obesity and diabetes. In addition, the role of lipid metabolism in cancer cell growth is receiving increased attention. Transcriptionally active SREBP molecules are unstable and rapidly degraded in a phosphorylation-dependent manner by Fbw7, a ubiquitin ligase that targets several cell cycle regulatory proteins for degradation. We have previously demonstrated that active SREBP1 is stabilized during mitosis. We have now delineated the mechanisms involved in the stabilization of SREBP1 in mitotic cells. This process is initiated by the phosphorylation of a specific serine residue in nuclear SREBP1 by the mitotic kinase Cdk1. The phosphorylation of this residue creates a docking site for a separate mitotic kinase, Plk1. Plk1 interacts with nuclear SREBP1 in mitotic cells and phosphorylates a number of residues in the C-terminal domain of the protein, including a threonine residue in close proximity of the Fbw7 docking site in SREBP1. The phosphorylation of these residues by Plk1 blocks the interaction between SREBP1 and Fbw7 and attenuates the Fbw7-dependent degradation of nuclear SREBP1 during cell division. Inactivation of SREBP1 results in a mitotic defect, suggesting that SREBP1 could regulate cell division. We propose that the mitotic phosphorylation and stabilization of nuclear SREBP1 during cell division provides a link between lipid metabolism and cell proliferation. Thus, the current study provides additional support for the emerging hypothesis that SREBP-dependent lipid metabolism may be important for cell growth.
Collapse
Affiliation(s)
| | - Johan Ericsson
- University College Dublin, School of Medicine and Medical Science, UCD Conway Institute, Dublin, Ireland
| |
Collapse
|
21
|
Gutteridge REA, Ndiaye MA, Liu X, Ahmad N. Plk1 Inhibitors in Cancer Therapy: From Laboratory to Clinics. Mol Cancer Ther 2016; 15:1427-35. [PMID: 27330107 PMCID: PMC4936921 DOI: 10.1158/1535-7163.mct-15-0897] [Citation(s) in RCA: 269] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 04/06/2016] [Indexed: 01/06/2023]
Abstract
Polo-like kinase 1 (Plk1) overexpression has been shown to occur in a wide range of tumors, prompting research and development of Plk1 inhibitors as a means of cancer treatment. This review discusses recent advances in the development of Plk1 inhibitors for cancer management. Plk1 inhibition has been shown to cause mitotic block and apoptosis of cells with higher mitotic index and therefore higher Plk1 expression. The potential of Plk1 inhibitors as cancer therapeutics has been widely investigated. However, a complete understanding of Plk1 biology/mechanism is yet to be fully achieved. Resistance to certain chemotherapeutic drugs has been linked to Plk1 overexpression, and Plk1-mediated mitotic events such as microtubule rearrangement have been found to reduce the efficacy of chemotherapeutic agents. The Plk1 inhibitor volasertib has shown considerable promise in clinical studies, having reached phase III trials. However, preclinical success with Plk1 inhibitors has not translated well into clinical success. In our view, combined therapies targeting other relevant pathways together with Plk1 may be vital to combat issues observed with monotherapy, especially resistance. In addition, research should also be directed toward understanding the mechanisms of Plk1 and designing additional next generations of specific, potent Plk1 inhibitors to target cancer. Mol Cancer Ther; 15(7); 1427-35. ©2016 AACR.
Collapse
Affiliation(s)
| | - Mary Ann Ndiaye
- Department of Dermatology, University of Wisconsin, Madison, Wisconsin
| | - Xiaoqi Liu
- Department of Biochemistry, Purdue University, West Lafayette, Indiana
| | - Nihal Ahmad
- Department of Dermatology, University of Wisconsin, Madison, Wisconsin. William S. Middleton Memorial VA Hospital, Madison, Wisconsin.
| |
Collapse
|
22
|
Cristóbal I, Rojo F, Madoz-Gúrpide J, García-Foncillas J. Cross Talk between Wnt/β-Catenin and CIP2A/Plk1 Signaling in Prostate Cancer: Promising Therapeutic Implications. Mol Cell Biol 2016; 36:1734-9. [PMID: 27090640 PMCID: PMC4907099 DOI: 10.1128/mcb.00130-16] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Aberrant activation of the Wnt/β-catenin pathway and polo-like kinase 1 (Plk1) overexpression represent two common events in prostate cancer with relevant functional implications. This minireview analyzes their potential therapeutic significance in prostate cancer based on their role as androgen receptor (AR) signaling regulators and the pivotal role of the tumor suppressor protein phosphatase 2A (PP2A) modulating these pathways.
Collapse
Affiliation(s)
- Ion Cristóbal
- Translational Oncology Division, Oncohealth Institute, IIS Fundación Jiménez Diaz, UAM, University Hospital Fundación Jiménez Diaz, Madrid, Spain
| | - Federico Rojo
- Pathology Department, IIS Fundación Jiménez Diaz, UAM, Madrid, Spain
| | | | - Jesús García-Foncillas
- Translational Oncology Division, Oncohealth Institute, IIS Fundación Jiménez Diaz, UAM, University Hospital Fundación Jiménez Diaz, Madrid, Spain
| |
Collapse
|
23
|
Fitzgerald KA, Malhotra M, Gooding M, Sallas F, Evans JC, Darcy R, O'Driscoll CM. A novel, anisamide-targeted cyclodextrin nanoformulation for siRNA delivery to prostate cancer cells expressing the sigma-1 receptor. Int J Pharm 2016; 499:131-145. [PMID: 26721726 DOI: 10.1016/j.ijpharm.2015.12.055] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 12/18/2015] [Accepted: 12/19/2015] [Indexed: 02/08/2023]
Abstract
Prostate cancer is a leading cause of cancer-related death in men and RNA interference (RNAi) has emerged as a potential therapeutic option. However, the absence of a safe and specific delivery vector remains a major obstacle to the clinical application of RNAi. Cyclodextrin derivatives are known to be efficient delivery systems with low toxicity in a variety of cell types. In this study, a cationic cyclodextrin derivative was synthesized to complex siRNA. The nanoparticle was then further modified by exploiting the ability of the β-cyclodextrin cavity to form an inclusion complex with the hydrophobic molecule adamantane. PEGylated adamantane derivatives were synthesized with and without the anisamide-targeting ligand on the terminal end of the PEG chain. Anisamide is known to bind specifically to the sigma receptor which is overexpressed on the surface of prostate cancer cells. The resulting nanocomplexes were slightly cationic and less than 300 nm in size. They successfully protected siRNA from serum-induced nuclease degradation and were non-toxic to prostate cancer cells. In addition, the targeted nanoparticles mediated high levels of siRNA cellular uptake and corresponding PLK1 gene knockdown in prostate cancer cells in vitro. To our knowledge, this is the first time that the ability of cyclodextrins to form inclusion complexes with adamantane derivatives has been exploited for the targeted delivery of siRNA to prostate cancer cells via the sigma receptor.
Collapse
Affiliation(s)
| | - Meenakshi Malhotra
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Ireland
| | - Matt Gooding
- Centre for Synthesis and Chemical Biology, University College Dublin, Ireland
| | - Florence Sallas
- Centre for Synthesis and Chemical Biology, University College Dublin, Ireland
| | - James C Evans
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Ireland
| | - Raphael Darcy
- Centre for Synthesis and Chemical Biology, University College Dublin, Ireland; School of Chemistry, University College Dublin, Ireland
| | | |
Collapse
|
24
|
Li J, Wang R, Schweickert PG, Karki A, Yang Y, Kong Y, Ahmad N, Konieczny SF, Liu X. Plk1 inhibition enhances the efficacy of gemcitabine in human pancreatic cancer. Cell Cycle 2016; 15:711-9. [PMID: 26890815 PMCID: PMC4845940 DOI: 10.1080/15384101.2016.1148838] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 01/19/2016] [Accepted: 01/20/2016] [Indexed: 01/21/2023] Open
Abstract
Gemcitabine is the standard-of-care for chemotherapy in patients with pancreatic adenocarcinoma and it can directly incorporate into DNA or inhibit ribonucleotide reductase to prevent DNA replication and, thus, tumor cell growth. Most pancreatic tumors, however, develop resistance to gemcitabine. Polo-like kinase 1 (Plk1), a critical regulator in many cell cycle events, is significantly elevated in human pancreatic cancer. In this study, we show that Plk1 is required for the G1/S transition and that inhibition of Plk1 significantly reduces the DNA synthesis rate in human pancreatic cancer cells. Furthermore, the combined effect of a specific Plk1 inhibitor GSK461364A with gemcitabine was examined. We show that inhibition of Plk1 significantly potentiates the anti-neoplastic activity of gemcitabine in both cultured pancreatic cancer cells and Panc1-derived orthotopic pancreatic cancer xenograft tumors. Overall, our study demonstrates that co-targeting Plk1 can significantly enhance the efficacy of gemcitabine, offering a promising new therapeutic option for the treatment of gemcitabine-resistant human pancreatic cancer.
Collapse
Affiliation(s)
- Jie Li
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | - Ruixin Wang
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | | | - Anju Karki
- Department of Biological Science, Purdue University, West Lafayette, IN, USA
| | - Yi Yang
- Department of Biological Science, Purdue University, West Lafayette, IN, USA
| | - Yifan Kong
- Department of Biological Science, Purdue University, West Lafayette, IN, USA
| | - Nihal Ahmad
- Department of Dermatology, University of Wisconsin, Madison, WI, USA
| | - Stephen F. Konieczny
- Department of Biological Science, Purdue University, West Lafayette, IN, USA
- Center for Cancer Research, Purdue University, West Lafayette, IN, USA
| | - Xiaoqi Liu
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
- Center for Cancer Research, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
25
|
Strebhardt K. Drugging Plk1: An attractive approach to inhibit androgen receptor signaling. Cell Cycle 2015; 14:2193-4. [PMID: 26030710 PMCID: PMC5242295 DOI: 10.1080/15384101.2015.1056611] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 05/19/2015] [Indexed: 01/17/2023] Open
Affiliation(s)
- Klaus Strebhardt
- Department of Gynecology; School of Medicine; Goethe University; Frankfurt, Germany
- German Cancer Consortium; Heidelberg, Germany
| |
Collapse
|