1
|
Xie H, Dan M, Cen Y, Ning J, Sun C, Zhu G, Feng S, Wang H, Pu J. AR expression-independent XRCC3 mediates DNA damage-induced p53/Bax signaling pathway activation against prostate cancer. J Cancer Res Clin Oncol 2024; 150:463. [PMID: 39414634 PMCID: PMC11485149 DOI: 10.1007/s00432-024-05989-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 10/05/2024] [Indexed: 10/18/2024]
Abstract
BACKGROUND Androgen deprivation therapy (ADT) resistance is closely associated with altered AR status. Aberrant AR expression is critical for the induction of ADT resistance, necessitating the identification of an anti-PCa target independent of AR expression. METHODS Transcriptomic data and clinical information of PRAD were obtained from TCGA database. Genes with PCa-related and AR expression-independent were screened by bioinformatics, and characterized by PPI and GO functional enrichment analyses. Candidate genes were locked by co-expression correlation and disease-free survival (DFS) analyses. A prognostic gene set was established using LASSO Cox regression algorithm. Cox proportional risk regression was performed to identify a key prognostic gene. Expression of the target protein in PCa tissues was verified by The Human Protein Atlas database. In vitro validation of cellular function and molecular mechanism by knockdown and overexpression of the target gene. RESULTS Two AR expression-independent genes (SLC43A1 and XRCC3) were available for the optimal prognostic model. This gene set effectively predicted PRAD patients' DFS at 1-, 3- and 5-year, where XRCC3 and tumor (T) stage were independent risk factors. XRCC3 was higher expressed in PRAD patients with T3-T4 stages and accompanied by poorer DFS. IHC staining also validated its higher expression in high-risk PCa tissues. In vitro experiments demonstrated that silencing XRCC3 significantly inhibited 22Rv1 and DU145 cell proliferation, migration and invasion, while promoted apoptosis. Further, silencing XRCC3 promoted DNA damage-induced p53/Bax signaling pathway activation, which was absent with overexpression. CONCLUSION Silencing XRCC3 exerts anti-PCa effects by promoting DNA damage-induced p53/Bax signaling pathway activation in an AR expression-independent manner.
Collapse
Affiliation(s)
- Hailong Xie
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
- Department of Urology, The Second Affiliated Hospital of Bengbu Medical College, Benbu, 233080, China
| | - Mingjiang Dan
- Department of Urology, Huiya Hospital of the First Affiliated Hospital of Sun Yat Sen University, Huizhou, 516081, China
| | - Yi Cen
- Guangzhou Medical University, Guangzhou, 511436, China
| | - Jing Ning
- Department of Urology, The Second Affiliated Hospital of Bengbu Medical College, Benbu, 233080, China
| | - Chong Sun
- Department of Urology, The Second Affiliated Hospital of Bengbu Medical College, Benbu, 233080, China
| | - Guangbin Zhu
- Department of Medical Imaging, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China
| | - Shourui Feng
- School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Haiyan Wang
- Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518100, China.
| | - Jinxian Pu
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
- Department of Urology, The Fourth Affiliated Hospital of Soochow University, Suzhou, 215000, China.
| |
Collapse
|
2
|
Sun W, Xu K, Li X, Qian P, Xu F, Zhang Y, Wang X, Xu Z, Ding J, Xu X, Wei X, Jiang Q, Xu Y. Insight into prostate cancer osteolytic metastasis by RelB coordination of IL-8 and S100A4. Clin Transl Med 2024; 14:e70058. [PMID: 39415352 PMCID: PMC11483529 DOI: 10.1002/ctm2.70058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 09/27/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND Although RANK-LRANK interaction is essential for osteoclastogenesis, the mechanisms by which cancer cells invade bone tissues and initiate osteolytic metastasis remain unclear. Here, we show that the hyperactivation of RelB fosters prostate cancer (PCa) osteolytic metastasis by coordinating interleukin-8 (IL-8) and calcium-binging protein A4 (S100A4). METHODS The factors promoting PCa bone metastasis were investigated in sera from PCa patients and tumour tissues derived from nude mice using immunohistochemical analysis and enzyme-linked immunosorbent assays (ELISA). Cell mobility and mineralization were quantified using BioStation CT and Osteolmage assay. The relative cistrome was investigated in advanced PCa cells by standard transcriptional analyses, including the luciferase reporter response, site-directed mutagenesis, and chromatin immunoprecipitation (ChIP) assay. PCa cell-initiated tumour formation, expansion, and bone metastasis were validated in mice using multiple approaches, including orthotopic, intraskeletal, and caudal arterial implantation models. RESULTS IL-8 and S100A4 correlated with patient Gleason scores and bone metastasis. RelB upregulated IL-8, facilitating androgen receptor (AR)-independent growth. RelB-Sp1 interaction enhanced epithelial-mesenchymal transition (EMT) by activating Snail and Twist. RelB-NFAT1c super-enhancer upregulated S100A4 in the organization of the cytoskeleton and bone metastasis. The RelB-IL-8-S100A4 signalling axis was confirmed to promote osteolytic metastasis in nude mice. CONCLUSION RelB-IL-8 reciprocally promoted EMT by activating inflammatory signalling and inactivating AR signalling. IL-8 is essential for provoking PCa metastasis but insufficient to drive bone metastasis. IL-8-S100A4 cooperation was necessary for metastatic cells to target the bone. HIGHLIGHTS RelB activates inflammatory signalling by upregulating IL-8 and suppressing AR. RelB upregulates S100A4 by cooperating with NFATC1. IL-8 boosts EMT by activating Snail 1 and Twist 1, and S100A4 exacerbates osteolytic metastasis via calcium consumption. RelB harnesses IL-8 and S100A4 to drive PCa osteolytic metastasis.
Collapse
Affiliation(s)
- Wenbo Sun
- Affiliated Eye HospitalNanjing Medical UniversityNanjingChina
- Affiliated Cancer HospitalNanjing Medical UniversityNanjingChina
- Department of Thoracic SurgeryThe First Affiliated HospitalNanjing Medical UniversityNanjingChina
| | - Kenny Xu
- College of Medicine Bowling Green CampusUniversity of KentuckyBowling GreenKentuckyUSA
| | - Xiao Li
- Affiliated Cancer HospitalNanjing Medical UniversityNanjingChina
| | - Peipei Qian
- Affiliated Eye HospitalNanjing Medical UniversityNanjingChina
- Affiliated Cancer HospitalNanjing Medical UniversityNanjingChina
| | - Fan Xu
- Affiliated Eye HospitalNanjing Medical UniversityNanjingChina
- Affiliated Cancer HospitalNanjing Medical UniversityNanjingChina
| | - Yanyan Zhang
- Affiliated Cancer HospitalNanjing Medical UniversityNanjingChina
| | - Xiumei Wang
- Affiliated Eye HospitalNanjing Medical UniversityNanjingChina
- Department of OncologyNanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Zhi Xu
- Affiliated Eye HospitalNanjing Medical UniversityNanjingChina
| | - Jiaji Ding
- Affiliated Cancer HospitalNanjing Medical UniversityNanjingChina
| | - Xinyu Xu
- Affiliated Cancer HospitalNanjing Medical UniversityNanjingChina
| | - Xiaowei Wei
- Department of OncologyNanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Qin Jiang
- Affiliated Eye HospitalNanjing Medical UniversityNanjingChina
| | - Yong Xu
- Affiliated Eye HospitalNanjing Medical UniversityNanjingChina
- Affiliated Cancer HospitalNanjing Medical UniversityNanjingChina
- Jiangsu Key Lab of Cancer BiomarkersPrevention, and TreatmentNanjing Medical UniversityNanjingChina
- Department of Toxicology and Cancer Biology & Markey Cancer CenterUniversity of KentuckyLexingtonKentuckyUSA
| |
Collapse
|
3
|
Tao J, Bian X, Zhou J, Zhang M. From microscopes to molecules: The evolution of prostate cancer diagnostics. Cytojournal 2024; 21:29. [PMID: 39391208 PMCID: PMC11464998 DOI: 10.25259/cytojournal_36_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/27/2024] [Indexed: 10/12/2024] Open
Abstract
In the ever-evolving landscape of oncology, the battle against prostate cancer (PCa) stands at a transformative juncture, propelled by the integration of molecular diagnostics into traditional cytopathological frameworks. This synthesis not only heralds a new epoch of precision medicine but also significantly enhances our understanding of the disease's genetic intricacies. Our comprehensive review navigates through the latest advancements in molecular biomarkers and their detection technologies, illuminating the potential these innovations hold for the clinical realm. With PCa persisting as one of the most common malignancies among men globally, the quest for early and precise diagnostic methods has never been more critical. The spotlight in this endeavor shines on the molecular diagnostics that reveal the genetic underpinnings of PCa, offering insights into its onset, progression, and resistance to conventional therapies. Among the genetic aberrations, the TMPRSS2-ERG fusion and mutations in genes such as phosphatase and tensin homolog (PTEN) and myelocytomatosis viral oncogene homolog (MYC) are identified as significant players in the disease's pathology, providing not only diagnostic markers but also potential therapeutic targets. This review underscores a multimodal diagnostic approach, merging molecular diagnostics with cytopathology, as a cornerstone in managing PCa effectively. This strategy promises a future where treatment is not only tailored to the individual's genetic makeup but also anticipates the disease's trajectory, offering hope for improved prognosis and quality of life for patients.
Collapse
Affiliation(s)
- Junyue Tao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xiaokang Bian
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jun Zhou
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Meng Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
4
|
Ji XZ, Qin X, Wang W, Wang L. A review of tanshinone compounds in prostate cancer treatment. Transl Androl Urol 2024; 13:1278-1287. [PMID: 39100845 PMCID: PMC11291418 DOI: 10.21037/tau-24-49] [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: 01/22/2024] [Accepted: 05/12/2024] [Indexed: 08/06/2024] Open
Abstract
Prostate cancer (PCa) is one of the most common malignant epithelial tumors in men worldwide. PCa patients are initially sensitive to chemotherapy, but patients in the advanced stages of PCa eventually develop resistance, leaving them with limited therapeutic options. Therefore, it is very important to screen new drugs for treating PCa. Salvia miltiorrhiza is a common Chinese herbal medicine used in some Asian countries. It has many functions and is widely used to treat a variety of diseases, including heart diseases and cancers. For the past few years, research has shown that liposoluble constituents of tanshinones (TANs), including cryptotanshinone, TAN IIA, dihydrotanshinone I, and TAN I, exhibit good anticancer activity in PCa. In this study, we review the progress of TAN compounds (cryptotanshinone, TAN IIA, dihydrotanshinone I, and TAN I) in treating PCa over the past decade. These compounds can act on the same molecular mechanisms, as they have a very similar structure; they are also found to work slightly differently in PCa. According to current studies, compared with other TAN compounds, TAN IIA appears to hold more potential for treating PCa. The toxicity, side effects or biodistribution of Salvia miltiorrhiza and these four TANs need to be confirmed with further research. Findings obtained in this study may provide important information for the potential clinical application of cryptotanshinone, TAN IIA, dihydrotanshinone I, and TAN I in the treatment of PCa.
Collapse
Affiliation(s)
- Xiao-Zhen Ji
- Department of Oncology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Xin Qin
- Department of Radiology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Wei Wang
- Department of Oncology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Lin Wang
- Department of Oncology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| |
Collapse
|
5
|
Su MC, Lee AM, Zhang W, Maeser D, Gruener RF, Deng Y, Huang RS. Computational Modeling to Identify Drugs Targeting Metastatic Castration-Resistant Prostate Cancer Characterized by Heightened Glycolysis. Pharmaceuticals (Basel) 2024; 17:569. [PMID: 38794139 PMCID: PMC11124089 DOI: 10.3390/ph17050569] [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: 03/29/2024] [Revised: 04/22/2024] [Accepted: 04/26/2024] [Indexed: 05/26/2024] Open
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) remains a deadly disease due to a lack of efficacious treatments. The reprogramming of cancer metabolism toward elevated glycolysis is a hallmark of mCRPC. Our goal is to identify therapeutics specifically associated with high glycolysis. Here, we established a computational framework to identify new pharmacological agents for mCRPC with heightened glycolysis activity under a tumor microenvironment, followed by in vitro validation. First, using our established computational tool, OncoPredict, we imputed the likelihood of drug responses to approximately 1900 agents in each mCRPC tumor from two large clinical patient cohorts. We selected drugs with predicted sensitivity highly correlated with glycolysis scores. In total, 77 drugs predicted to be more sensitive in high glycolysis mCRPC tumors were identified. These drugs represent diverse mechanisms of action. Three of the candidates, ivermectin, CNF2024, and P276-00, were selected for subsequent vitro validation based on the highest measured drug responses associated with glycolysis/OXPHOS in pan-cancer cell lines. By decreasing the input glucose level in culture media to mimic the mCRPC tumor microenvironments, we induced a high-glycolysis condition in PC3 cells and validated the projected higher sensitivity of all three drugs under this condition (p < 0.0001 for all drugs). For biomarker discovery, ivermectin and P276-00 were predicted to be more sensitive to mCRPC tumors with low androgen receptor activities and high glycolysis activities (AR(low)Gly(high)). In addition, we integrated a protein-protein interaction network and topological methods to identify biomarkers for these drug candidates. EEF1B2 and CCNA2 were identified as key biomarkers for ivermectin and CNF2024, respectively, through multiple independent biomarker nomination pipelines. In conclusion, this study offers new efficacious therapeutics beyond traditional androgen-deprivation therapies by precisely targeting mCRPC with high glycolysis.
Collapse
Affiliation(s)
- Mei-Chi Su
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (M.-C.S.); (A.M.L.); (R.F.G.)
| | - Adam M. Lee
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (M.-C.S.); (A.M.L.); (R.F.G.)
| | - Weijie Zhang
- Bioinformatics and Computational Biology, University of Minnesota, Minneapolis, MN 55455, USA; (W.Z.); (D.M.)
| | - Danielle Maeser
- Bioinformatics and Computational Biology, University of Minnesota, Minneapolis, MN 55455, USA; (W.Z.); (D.M.)
| | - Robert F. Gruener
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (M.-C.S.); (A.M.L.); (R.F.G.)
| | - Yibin Deng
- Department of Urology, Masonic Cancer Center, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - R. Stephanie Huang
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (M.-C.S.); (A.M.L.); (R.F.G.)
- Bioinformatics and Computational Biology, University of Minnesota, Minneapolis, MN 55455, USA; (W.Z.); (D.M.)
| |
Collapse
|
6
|
Granata I, Barboro P. Identification of Molecular Markers Associated with Prostate Cancer Subtypes: An Integrative Bioinformatics Approach. Biomolecules 2024; 14:87. [PMID: 38254687 PMCID: PMC10813078 DOI: 10.3390/biom14010087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/24/2024] Open
Abstract
Prostate cancer (PCa) is characterised by androgen dependency. Unfortunately, under anti-androgen treatment pressure, castration-resistant prostate cancer (CRPC) emerges, characterised by heterogeneous cell populations that, over time, lead to the development of different androgen-dependent or -independent phenotypes. Despite important advances in therapeutic strategies, CRPC remains incurable. Context-specific essential genes represent valuable candidates for targeted anti-cancer therapies. Through the investigation of gene and protein annotations and the integration of published transcriptomic data, we identified two consensus lists to stratify PCa patients' risk and discriminate CRPC phenotypes based on androgen receptor activity. ROC and Kaplan-Meier survival analyses were used for gene set validation in independent datasets. We further evaluated these genes for their association with cancer dependency. The deregulated expression of the PCa-related genes was associated with overall and disease-specific survival, metastasis and/or high recurrence risk, while the CRPC-related genes clearly discriminated between adeno and neuroendocrine phenotypes. Some of the genes showed context-specific essentiality. We further identified candidate drugs through a computational repositioning approach for targeting these genes and treating lethal variants of PCa. This work provides a proof-of-concept for the use of an integrative approach to identify candidate biomarkers involved in PCa progression and CRPC pathogenesis within the goal of precision medicine.
Collapse
Affiliation(s)
- Ilaria Granata
- High Performance Computing and Networking Institute (ICAR), National Council of Research (CNR), Via Pietro Castellino 111, 80131 Naples, Italy
| | - Paola Barboro
- Proteomic and Mass Spectrometry Unit, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genoa, Italy;
| |
Collapse
|
7
|
Stangis MM, Colah AN, McLean DT, Halberg RB, Collier LS, Ricke WA. Potential roles of FGF5 as a candidate therapeutic target in prostate cancer. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2023; 11:452-466. [PMID: 38148937 PMCID: PMC10749387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/13/2023] [Indexed: 12/28/2023]
Abstract
Fibroblast growth factor (FGF) is a secreted ligand that is widely expressed in embryonic tissues but its expression decreases with age. In the developing prostate, FGF5 has been proposed to interact with the Hedgehog (Hh) signaling pathway to guide mitogenic processes. In the adult prostate, the FGF/FGFR signaling axis has been implicated in prostate carcinogenesis, but focused studies on FGF5 functions in the prostate are limited. Functional studies completed in other cancer models point towards FGF5 overexpression as an oncogenic driver associated with stemness, metastatic potential, proliferative capacity, and increased tumor grade. In this review, we explore the significance of FGF5 as a therapeutic target in prostate cancer (PCa) and other malignancies; and we introduce a potential route of investigation to link FGF5 to benign prostatic hyperplasia (BPH). PCa and BPH are two primary contributors to the disease burden of the aging male population and have severe implications on quality of life, psychological wellbeing, and survival. The development of new FGF5 inhibitors could potentially alleviate the health burden of PCa and BPH in the aging male population.
Collapse
Affiliation(s)
- Mary M Stangis
- Department of Urology, University of Wisconsin-MadisonMadison, WI, USA
- Department of Oncology, McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public HealthMadison, WI, USA
| | - Avan N Colah
- Department of Urology, University of Wisconsin-MadisonMadison, WI, USA
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-MadisonMadison, WI, USA
| | - Dalton T McLean
- Department of Urology, University of Wisconsin-MadisonMadison, WI, USA
| | - Richard B Halberg
- Department of Oncology, McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public HealthMadison, WI, USA
- Carbone Cancer Center, University of Wisconsin-MadisonMadison, WI, USA
| | - Lara S Collier
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-MadisonMadison, WI, USA
| | - William A Ricke
- Department of Urology, University of Wisconsin-MadisonMadison, WI, USA
- Carbone Cancer Center, University of Wisconsin-MadisonMadison, WI, USA
| |
Collapse
|
8
|
Feng K, Liu C, Wang W, Kong P, Tao Z, Liu W. Emerging proteins involved in castration‑resistant prostate cancer via the AR‑dependent and AR‑independent pathways (Review). Int J Oncol 2023; 63:127. [PMID: 37732538 PMCID: PMC10609492 DOI: 10.3892/ijo.2023.5575] [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: 06/26/2023] [Accepted: 09/06/2023] [Indexed: 09/22/2023] Open
Abstract
Despite achieving optimal initial responses to androgen deprivation therapy, most patients with prostate cancer eventually progress to a poor prognosis state known as castration‑resistant prostate cancer (CRPC). Currently, there is a notable absence of reliable early warning biomarkers and effective treatment strategies for these patients. Although androgen receptor (AR)‑independent pathways have been discovered and acknowledged in recent years, the AR signaling pathway continues to play a pivotal role in the progression of CRPC. The present review focuses on newly identified proteins within human CRPC tissues. These proteins encompass both those involved in AR‑dependent and AR‑independent pathways. Specifically, the present review provides an in‑depth summary and analysis of the emerging proteins within AR bypass pathways. Furthermore, the significance of these proteins as potential biomarkers and therapeutic targets for treating CRPC is discussed. Therefore, the present review offers valuable theoretical insights and clinical perspectives to comprehensively enhance the understanding of CRPC.
Collapse
Affiliation(s)
- Kangle Feng
- Department of Blood Transfusion, Shaoxing Central Hospital, Shaoxing, Zhejiang 312030, P.R. China
- Department of Laboratory Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Chunhua Liu
- Department of Blood Transfusion, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Weixi Wang
- Department of Laboratory Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Piaoping Kong
- Department of Laboratory Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Zhihua Tao
- Department of Laboratory Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Weiwei Liu
- Department of Laboratory Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| |
Collapse
|
9
|
Han X, Zhao L, Xiang W, Wang M, Miao B, Qin C, McEachern D, Lu J, Wang Y, Metwally H, Kirchhoff P, Wang L, Matvekas A, Takyi-Williams J, Wen B, Sun D, Ator M, Mckean R, Wang S. Discovery of ARD-2051 as a Potent and Orally Efficacious Proteolysis Targeting Chimera (PROTAC) Degrader of Androgen Receptor for the Treatment of Advanced Prostate Cancer. J Med Chem 2023; 66:8822-8843. [PMID: 37382562 PMCID: PMC10568492 DOI: 10.1021/acs.jmedchem.3c00405] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
We report the discovery of ARD-2051 as a potent and orally efficacious androgen receptor (AR) proteolysis-targeting chimera degrader. ARD-2051 achieves DC50 values of 0.6 nM and Dmax >90% in inducing AR protein degradation in both the LNCaP and VCaP prostate cancer cell lines, potently and effectively suppresses AR-regulated genes, and inhibits cancer cell growth. ARD-2051 achieves a good oral bioavailability and pharmacokinetic profile in mouse, rat, and dog. A single oral dose of ARD-2051 strongly reduces AR protein and suppresses AR-regulated gene expression in the VCaP xenograft tumor tissue in mice. Oral administration of ARD-2051 effectively inhibits VCaP tumor growth and causes no signs of toxicity in mice. ARD-2051 is a promising AR degrader for advanced preclinical development for the treatment of AR+ human cancers.
Collapse
Affiliation(s)
- Xin Han
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Lijie Zhao
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Weiguo Xiang
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Mi Wang
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Bukeyan Miao
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Chong Qin
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Donna McEachern
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jianfeng Lu
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Yu Wang
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Hoda Metwally
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Paul Kirchhoff
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Lu Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Aleksas Matvekas
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - John Takyi-Williams
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Bo Wen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Mark Ator
- Oncopia Therapeutics Inc, 2 West Liberty Blvd. Malvern, PA 19355 USA
| | - Robert Mckean
- Oncopia Therapeutics Inc, 2 West Liberty Blvd. Malvern, PA 19355 USA
| | - Shaomeng Wang
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
10
|
Satapathy S, Sahoo RK, Bal C. [ 177Lu]Lu-PSMA-Radioligand Therapy Efficacy Outcomes in Taxane-Naïve Versus Taxane-Treated Patients with Metastatic Castration-Resistant Prostate Cancer: A Systematic Review and Metaanalysis. J Nucl Med 2023:jnumed.123.265414. [PMID: 37169534 DOI: 10.2967/jnumed.123.265414] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/10/2023] [Indexed: 05/13/2023] Open
Abstract
Radioligand therapy (RLT) with 177Lu-prostate-specific membrane antigen (PSMA) inhibitors ([177Lu]Lu-PSMA) is currently approved for patients with metastatic castration-resistant prostate cancer (mCRPC) after progression with at least 1 taxane and 1 androgen-receptor-pathway inhibitor. However, the impact of prior chemotherapy on [177Lu]Lu-PSMA-RLT outcomes is debatable, with various studies showing inconsistent results. This study was conducted to precisely evaluate the impact of prior taxane chemotherapy on response and survival outcomes in mCRPC patients after [177Lu]Lu-PSMA-RLT. Methods: This review followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Searches in PubMed, Scopus, and Embase were made using relevant key words, and articles up to December 2022 were included. The endpoints included prostate-specific antigen (PSA) response rate (RR), progression-free survival, and overall survival (OS). Individual patient data were pooled when feasible. Univariate odds ratios (ORs) and hazard ratios (HRs) were extracted from the individual articles, and pooled estimates and 95% CIs were generated using metaanalysis. Results: Thirteen articles comprising 2,068 patients were included. In 6 articles (553 patients), taxane-naïve patients had significantly better odds of biochemical response after [177Lu]Lu-PSMA-RLT (pooled OR, 1.82; 95% CI, 1.21-2.71). Individual patient data metaanalysis for PSA RRs in 3 articles revealed a significantly higher PSA RR in the taxane-naïve versus taxane-treated patients (57.1% vs. 39.5%; difference, 17.6%; 95% CI, 5.6%-28.9%). Further, taxane-naïve status was also a predictor of significantly better progression-free survival (5 articles; 1,027 patients; pooled HR, 0.60; 95% CI, 0.51-0.69) and OS (8 articles; 1,594 patients; pooled HR, 0.54; 95% CI, 0.43-0.68) after [177Lu]Lu-PSMA-RLT. There was no evidence of publication bias. Conclusion: mCRPC patients with no prior taxanes had significantly better outcomes after [177Lu]Lu-PSMA-RLT than did taxane-treated patients. Further trials evaluating [177Lu]Lu-PSMA-RLT in the taxane-naïve setting are now required.
Collapse
Affiliation(s)
- Swayamjeet Satapathy
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India; and
| | - Ranjit K Sahoo
- Department of Medical Oncology, B.R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Chandrasekhar Bal
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India; and
| |
Collapse
|
11
|
Yang Z, Gao Y, He K, Sui X, Chen J, Wang T, Chen M, Wang Z, Yi J, Zhao L. Voluntarily wheel running inhibits the growth of CRPC xenograft by inhibiting HMGB1 in mice. Exp Gerontol 2023; 174:112118. [PMID: 36758649 DOI: 10.1016/j.exger.2023.112118] [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: 12/21/2022] [Revised: 01/17/2023] [Accepted: 02/05/2023] [Indexed: 02/11/2023]
Abstract
INTRODUCTION Exercise has been proved to reduce the risk of recurrence and mortality of cancer. Emerging evidence indicated that exercise may regulate both systematical and local metabolism, immunity and other ways. Although the role of exercise in inhibiting castration-resistant prostate cancer is well established, the underlying mechanism remains unclear. METHOD Twenty C57BL/6 male mice were used to construct CRPC xenograft models and randomly divided into exercise group (n = 10) and control group (n = 10). After exercised with voluntarily wheel running for 21 days, the mice were sacrificed and the tumor tissues and serum were collected. TUNEL staining was used to detect the apoptosis of tumor cells. The expression of PI3K signal pathway and apoptosis related proteins were detected by Western blot. The expression of AR and HMGB1 were examined by Western blot and Immunohistochemical staining. IFN-γ, TNF-α, TGF-β, IL-4, IL-6, IL-10 in serum was examined using ELISA kits. RESULTS Voluntarily wheel running inhibited the growth of CRPC xenografts, inhibited the proliferation of tumor cells and promoted the apoptosis of tumor cells. HMGB1 levels in serum and tumor tissues were significantly reduced after exercise, which enhanced local immunity by inducing more leukocyte infiltration and inhibited systemic inflammatory response by regulating cytokines. CONCLUSION Voluntary wheel running can down-regulate the expression of HMGB1 in serum and transplanted tumor tissues, inhibit proliferation and promote apoptosis of tumor cells, enhance immune cell infiltration and systemic inflammatory response, and regulate local anti-tumor effects in tumor microenvironment.
Collapse
Affiliation(s)
- Zhaoyun Yang
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun 130021, China
| | - Yan Gao
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun 130021, China
| | - Kang He
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun 130021, China
| | - Xin Sui
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun 130021, China
| | - Junyu Chen
- Department of Gynecology and Obstetrics, the Second Hospital of Jilin University, Changchun 130041, China
| | - Taiwei Wang
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun 130021, China
| | - Mengmeng Chen
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun 130021, China
| | - Zeyu Wang
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun 130021, China
| | - Jiang Yi
- Department of Rehabilitation, The Second Hospital of Jilin University, Changchun 130041, China.
| | - Lijing Zhao
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun 130021, China.
| |
Collapse
|
12
|
Shahrokhi P, Emami-Ardekani A, Karamzade-Ziarati N. SSTR-based theranostics in neuroendocrine prostate cancer (NEPC). Clin Transl Imaging 2022. [DOI: 10.1007/s40336-022-00535-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
13
|
Advances in the Current Understanding of the Mechanisms Governing the Acquisition of Castration-Resistant Prostate Cancer. Cancers (Basel) 2022; 14:cancers14153744. [PMID: 35954408 PMCID: PMC9367587 DOI: 10.3390/cancers14153744] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 07/29/2022] [Indexed: 11/17/2022] Open
Abstract
Despite aggressive treatment and androgen-deprivation therapy, most prostate cancer patients ultimately develop castration-resistant prostate cancer (CRPC), which is associated with high mortality rates. However, the mechanisms governing the development of CRPC are poorly understood, and androgen receptor (AR) signaling has been shown to be important in CRPC through AR gene mutations, gene overexpression, co-regulatory factors, AR shear variants, and androgen resynthesis. A growing number of non-AR pathways have also been shown to influence the CRPC progression, including the Wnt and Hh pathways. Moreover, non-coding RNAs have been identified as important regulators of the CRPC pathogenesis. The present review provides an overview of the relevant literature pertaining to the mechanisms governing the molecular acquisition of castration resistance in prostate cancer, providing a foundation for future, targeted therapeutic efforts.
Collapse
|
14
|
Ji X, Liu K, Li Q, Shen Q, Han F, Ye Q, Zheng C. A Mini-Review of Flavone Isomers Apigenin and Genistein in Prostate Cancer Treatment. Front Pharmacol 2022; 13:851589. [PMID: 35359832 PMCID: PMC8962830 DOI: 10.3389/fphar.2022.851589] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 01/20/2022] [Indexed: 12/19/2022] Open
Abstract
The initial responses to standard chemotherapies among prostate cancer (PCa) patients are usually significant, while most of them will finally develop drug resistance, rendering them with limited therapies. To discover new regimens for the treatment of PCa including resistant PCa, natural products, the richest source of bioactive compounds, can serve as a library for screening and identifying promising candidates, and flavones such as apigenin and genistein have been used in lab and clinical trials for treating PCa over decades. In this mini-review, we take a look into the progress of apigenin and genistein, which are isomers, in treating PCa in the past decade. While possessing very similar structure, these two isomers can both target the same signaling pathways; they also are found to work differently in PCa cells. Given that more combinations are being developed and tested, genistein appears to be the more promising option to be approved. The anticancer efficacies of these two flavones can be confirmed by in-vitro and in-vivo studies, and their applications remain to be validated in clinical trials. Information gained in this work may provide important information for new drug development and the potential application of apigenin and genistein in treating PCa.
Collapse
Affiliation(s)
- Xiaozhen Ji
- Hainan General Hospital and Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Kai Liu
- Hainan General Hospital and Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Qingyue Li
- Hainan General Hospital and Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Qun Shen
- Hainan General Hospital and Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Fangxuan Han
- Hainan General Hospital and Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Qingmei Ye
- Hainan General Hospital and Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
- Key Laboratory of Tropical Medicinal Plant Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, China
- Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, China
- *Correspondence: Qingmei Ye, ; Caijuan Zheng,
| | - Caijuan Zheng
- Key Laboratory of Tropical Medicinal Plant Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, China
- Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, China
- *Correspondence: Qingmei Ye, ; Caijuan Zheng,
| |
Collapse
|
15
|
Monga J, Adrianto I, Rogers C, Gadgeel S, Chitale D, Alumkal JJ, Beltran H, Zoubeidi A, Ghosh J. Tribbles 2 pseudokinase confers enzalutamide resistance in prostate cancer by promoting lineage plasticity. J Biol Chem 2022; 298:101556. [PMID: 34973338 PMCID: PMC8800106 DOI: 10.1016/j.jbc.2021.101556] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/10/2021] [Accepted: 12/12/2021] [Indexed: 02/03/2023] Open
Abstract
Enzalutamide, a second-generation antiandrogen, is commonly prescribed for the therapy of advanced prostate cancer, but enzalutamide-resistant, lethal, or incurable disease invariably develops. To understand the molecular mechanism(s) behind enzalutamide resistance, here, we comprehensively analyzed a range of prostate tumors and clinically relevant models by gene expression array, immunohistochemistry, and Western blot, which revealed that enzalutamide-resistant prostate cancer cells and tumors overexpress the pseudokinase, Tribbles 2 (TRIB2). Inhibition of TRIB2 decreases the viability of enzalutamide-resistant prostate cancer cells, suggesting a critical role of TRIB2 in these cells. Moreover, the overexpression of TRIB2 confers resistance in prostate cancer cells to clinically relevant doses of enzalutamide, and this resistance is lost upon inhibition of TRIB2. Interestingly, we found that TRIB2 downregulates the luminal markers androgen receptor and cytokeratin 8 in prostate cancer cells but upregulates the neuronal transcription factor BRN2 (Brain-2) and the stemness factor SOX2 (SRY-box 2) to induce neuroendocrine characteristics. Finally, we show that inhibition of either TRIB2 or its downstream targets, BRN2 or SOX2, resensitizes resistant prostate cancer cells to enzalutamide. Thus, TRIB2 emerges as a potential new regulator of transdifferentiation that confers enzalutamide resistance in prostate cancer cells via a mechanism involving increased cellular plasticity and lineage switching.
Collapse
Affiliation(s)
- Jitender Monga
- Vattikuti Urology Institute, Henry Ford Health System, Detroit, Michigan, USA
| | - Indra Adrianto
- Public Health Sciences, Henry Ford Health System, Detroit, Michigan, USA
| | - Craig Rogers
- Vattikuti Urology Institute, Henry Ford Health System, Detroit, Michigan, USA; Henry Ford Cancer Institute, Henry Ford Health System, Detroit, Michigan, USA
| | - Shirish Gadgeel
- Henry Ford Cancer Institute, Henry Ford Health System, Detroit, Michigan, USA
| | - Dhananjay Chitale
- Henry Ford Cancer Institute, Henry Ford Health System, Detroit, Michigan, USA; Department of Pathology, Henry Ford Health System, Detroit, Michigan, USA
| | - Joshi J Alumkal
- Department of Internal Medicine, Univeristy of Michigan Rogel Cancer Center, Ann Arbor, Michigan, USA
| | - Himisha Beltran
- Department of Medical Oncology, Dana Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Amina Zoubeidi
- Department of Urologic Sciences, University of British Columbia and The Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Jagadananda Ghosh
- Vattikuti Urology Institute, Henry Ford Health System, Detroit, Michigan, USA; Henry Ford Cancer Institute, Henry Ford Health System, Detroit, Michigan, USA.
| |
Collapse
|
16
|
Impact of Progressive Site-Directed Therapy in Oligometastatic Castration-Resistant Prostate Cancer on Subsequent Treatment Response. Cancers (Basel) 2022; 14:cancers14030567. [PMID: 35158833 PMCID: PMC8833545 DOI: 10.3390/cancers14030567] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Local treatment for oligometastatic hormone-naive prostate cancer has been shown to be effective in phase II trials. As for the efficacy of targeted therapy for oligometastatic castration-resistant prostate cancer, the results of the phase trial are not yet available, but the number of reports showing efficacy by retrospective analysis is increasing. Progressive site-directed therapy has been shown to delay the next intervention and prolong progression-free survival, but its impact on subsequent treatment efficacy and contribution to overall survival has not been reported. The purpose of this retrospective study is to evaluate the impact of progressive site-directed therapy for oligometastatic castration-resistant prostate cancer on the subsequent treatment outcomes. We found that progressive site-directed therapy was associated with better response to subsequent androgen receptor axis-targeted drugs and better overall survival. Progressive site-directed therapy for oligometastatic castration-resistant prostate cancer may improve subsequent oncological outcomes. Abstract The purpose of this study was to evaluate the impact of progressive site-directed therapy (PSDT) for oligometastatic castration-resistant prostate cancer (OM-CRPC) on the efficacy of subsequent androgen receptor axis-targeted (ARAT) drugs, and to demonstrate the possibility of prolonging overall survival (OS). We performed a retrospective analysis of 15 OM-CRPC patients who underwent PSDT and subsequently received first-line ARAT drugs (PSDT group) and 13 OM-CRPC patients who were treated with first-line ARAT drugs without PSDT (non-PSDT group). PSDT was performed with the intention of treating all progressing sites detected by whole-body diffusion-weighted MRI with radiotherapy. Thirteen patients (86.7%) treated with PSDT had a decrease in PSA levels, which was at least 50% in 10 (66.7%) patients. The median PSA progression-free survival (PFS) for PSDT was 7.4 months. The median PSA-PFS for ARAT was 27.2 months in patients in the PSDT group and 11.7 months in the non-PSDT group, with a significant difference between the two groups (hazard ratio [HR], 0.28; p = 0.010). The median OS was not reached in the PSDT group and was significantly longer than 44.5 months in the non-PSDT group (HR, 0.11; p = 0.014). In OM-CRPC, PSDT may improve the efficacy of subsequent ARAT and OS.
Collapse
|
17
|
Hu CY, Wu KY, Lin TY, Chen CC. The Crosstalk of Long Non-Coding RNA and MicroRNA in Castration-Resistant and Neuroendocrine Prostate Cancer: Their Interaction and Clinical Importance. Int J Mol Sci 2021; 23:ijms23010392. [PMID: 35008817 PMCID: PMC8745162 DOI: 10.3390/ijms23010392] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/02/2021] [Accepted: 12/28/2021] [Indexed: 12/18/2022] Open
Abstract
Prostate cancer is featured by its heterogeneous nature, which indicates a different prognosis. Castration-resistant prostate cancer (CRPC) is a hallmark of the treatment-refractory stage, and the median survival of patients is only within two years. Neuroendocrine prostate cancer (NEPC) is an aggressive variant that arises from de novo presentation of small cell carcinoma or treatment-related transformation with a median survival of 1–2 years from the time of diagnosis. The epigenetic regulators, such as long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), have been proven involved in multiple pathologic mechanisms of CRPC and NEPC. LncRNAs can act as competing endogenous RNAs to sponge miRNAs that would inhibit the expression of their targets. After that, miRNAs interact with the 3’ untranslated region (UTR) of target mRNAs to repress the step of translation. These interactions may modulate gene expression and influence cancer development and progression. Otherwise, epigenetic regulators and genetic mutation also promote neuroendocrine differentiation and cancer stem-like cell formation. This step may induce neuroendocrine prostate cancer development. This review aims to provide an integrated, synthesized overview under current evidence to elucidate the crosstalk of lncRNAs with miRNAs and their influence on castration resistance or neuroendocrine differentiation of prostate cancer. Notably, we also discuss the mechanisms of lncRNA–miRNA interaction in androgen receptor-independent prostate cancer, such as growth factors, oncogenic signaling pathways, cell cycle dysregulation, and cytokines or other transmembrane proteins. Conclusively, we underscore the potential of these communications as potential therapeutic targets in the future.
Collapse
Affiliation(s)
- Che-Yuan Hu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan;
- Department of Urology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan;
| | - Kuan-Yu Wu
- Department of Urology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan;
| | - Tsung-Yen Lin
- Department of Urology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan;
- Division of Urology, Department of Surgery, Dou-Liou Branch, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Yunlin 640, Taiwan
- Correspondence: (T.-Y.L.); (C.-C.C.); Tel.: +886-6235-3535 (ext. 5251) (T.-Y.L.); +886-5276-5041 (ext. 7521) (C.-C.C.)
| | - Chien-Chin Chen
- Department of Pathology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 600, Taiwan
- Department of Cosmetic Science, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan
- Correspondence: (T.-Y.L.); (C.-C.C.); Tel.: +886-6235-3535 (ext. 5251) (T.-Y.L.); +886-5276-5041 (ext. 7521) (C.-C.C.)
| |
Collapse
|