1
|
Liang H, Zhou B, Li P, Zhang X, Zhang S, Zhang Y, Yao S, Qu S, Chen J. Stemness regulation in prostate cancer: prostate cancer stem cells and targeted therapy. Ann Med 2025; 57:2442067. [PMID: 39711287 DOI: 10.1080/07853890.2024.2442067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 11/07/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Increasing evidence indicates that cancer stem cells (CSCs) and cancer stem-like cells form a special subpopulation of cells that are ubiquitous in tumors. These cells exhibit similar characteristics to those of normal stem cells in tissues; moreover, they are capable of self-renewal and differentiation, as well as high tumorigenicity and drug resistance. In prostate cancer (PCa), it is difficult to kill these cells using androgen signaling inhibitors and chemotherapy drugs. Consequently, the residual prostate cancer stem cells (PCSCs) mediate tumor recurrence and progression. OBJECTIVE This review aims to provide a comprehensive and up-to-date overview of PCSCs, with a particular emphasis on potential therapeutic strategies targeting these cells. METHODS After searching in PubMed and Embase databases using 'prostate cancer' and 'cancer stem cells' as keywords, studies related were compiled and examined. RESULTS In this review, we detail the origin and characteristics of PCSCs, introduce the regulatory pathways closely related to CSC survival and stemness maintenance, and discuss the link between epithelial-mesenchymal transition, tumor microenvironment and tumor stemness. Furthermore, we introduce the currently available therapeutic strategies targeting CSCs, including signaling pathway inhibitors, anti-apoptotic protein inhibitors, microRNAs, nanomedicine, and immunotherapy. Lastly, we summarize the limitations of current CSC research and mention future research directions. CONCLUSION A deeper understanding of the regulatory network and molecular markers of PCSCs could facilitate the development of novel therapeutic strategies targeting these cells. Previous preclinical studies have demonstrated the potential of this treatment approach. In the future, this may offer alternative treatment options for PCa patients.
Collapse
Affiliation(s)
- Hao Liang
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| | - Bin Zhou
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Peixin Li
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoyi Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Shijie Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Yaozhong Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Shengwen Yao
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Sifeng Qu
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| | - Jun Chen
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| |
Collapse
|
2
|
Yu YZ, Xie X, Cai MP, Hong YY, Ren YZ, Kang X, Yan HC, Xiong Y, Chen H, Wu XC, Luo DS, Zhao SC. Identification of pyrimidine metabolism-based molecular subtypes and prognostic signature to predict immune landscape and guide clinical treatment in prostate cancer. Ann Med 2025; 57:2449584. [PMID: 39803822 PMCID: PMC11731156 DOI: 10.1080/07853890.2025.2449584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 09/03/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND We previously described the enrichment of plasma exosome metabolites in CRPC, PCa, and TFC cohorts, and found significant differences in pyrimidine metabolites. The PMGs is associated with the clinical prognosis of several cancers, but its biological role in PCa is still unclear. METHODS This study extracted 98 reliable PMGs, and analyzed their somatic mutations, expression levels, and prognostic significance. Unsupervised clustering was applied to classify patients with PCa into clusters based on six PMGs that were related to the prognosis of PCa. The TME, gene mutations, and immune escape ability were compared among the clusters. A scoring algorithm based on prognostic PMGs, referred to as the PMGscore, was developed. TK1 was identified and the biological functions of TK1 were determined using loss-of-function experiments. RNA sequencing was subsequently performed to determine the molecules associated with the underlying mechanisms of TK1 function. RESULTS In total, six out of 98 PMGs simultaneously exhibited differential expression in PCa and were correlated with BCR. Patients were clustered into two clusters according to the expression levels of these six PMGs, which reflected distinct clinical outcomes and immune cell infiltration characteristics. Clinical features, tumor prognosis, and functional annotation were analyzed. Subsequently, we constructed a prognostic signature using these six PMGs. In combination with other clinical traits, we found that the six PMGs' prognostic signature was an independent prognostic factor for patients with PCa. Finally, we found that the expression of TK1 was higher in CRPC tissues than in PCa tissues in three GEO datasets. The results indicated that TK1 promotes the growth and metastasis of PCa cells. CONCLUSIONS We provide evidence for a PMG signature for PCa patients to accurately predict clinical prognosis. TK1 plays crucial roles in the progression of PCa cells and can be used as a potential therapeutic target for CRPC.
Collapse
Affiliation(s)
- Yu-Zhong Yu
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiao Xie
- Department of Urology, Pingxiang People’s Hospital, Pingxiang, China
| | - Mao-Ping Cai
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Ya-Ying Hong
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yang-Zi Ren
- Department of Oncology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xi Kang
- Department of Urology, Pingxiang People’s Hospital, Pingxiang, China
| | - Hai-Chen Yan
- Department of Urology, Pingxiang People’s Hospital, Pingxiang, China
| | - Yang Xiong
- Department of Urology, Pingxiang People’s Hospital, Pingxiang, China
| | - Hong Chen
- Luoyang Key Laboratory of Organic Functional Molecules, College of Food and Drug, Luoyang Normal University, Luoyang, China
| | - Xing-Cheng Wu
- Department of Urology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Dao-Sheng Luo
- Department of Urology, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People’s Hospital), Dongguan, China
| | - Shan-Chao Zhao
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
3
|
Zhao Y, Ma Y, Leng Q, Zhang Q, Li Y, Ji M, Yang H, Li X, Jia G, Li Z, Liu H, Zhang J. Engineered cell nanovesicle antagonists for androgen deprivation therapy of melanoma. J Colloid Interface Sci 2025; 691:137468. [PMID: 40179551 DOI: 10.1016/j.jcis.2025.137468] [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: 02/22/2025] [Revised: 03/24/2025] [Accepted: 03/28/2025] [Indexed: 04/05/2025]
Abstract
Epidemiological studies on melanoma have revealed significant gender disparities, with the incidence and mortality rates being higher in males than in females. Recent studies indicate that androgen contributing to T cell exhaustion and promoting cancer cell proliferation. While clinical androgen deprivation therapies (ADT),particularly the use of androgen receptor (AR) antagonists to block AR signaling, has been employed in clinical settings to reduce androgen levels, antiandrogen drugs often encounter challenges such as poor targeting and selectivity, increased toxicity, low stability, short half-life and the emergence of drug resistance. Here, we establish a nanoantagonists for efficient AR signaling blockade by arming antigen-activated dendritic cells (DCs) nanovesicles with AR antibodies (aAR-NVOVA). This innovative approach demonstrates dual therapeutic efficacy: aAR-NVOVA effectively disrupts androgen-AR interactions in both melanoma cells and T cells, simultaneously inhibiting tumor proliferation and reversing T cell exhaustion. Furthermore, aAR-NVOVA retains the inherent immunostimulatory properties of DCs, facilitating T cell activation and enhancing cytotoxic T lymphocyte infiltration within tumor tissues. As a result, a synergistic effect has been observed in boosting T cell-based immunotherapy by simultaneously enhancing T cell activity and reducing its exhaustion. Our study using aAR-NVOVA to antagonize androgen effects offers a promising new strategy for enhancing melanoma immunotherapy.
Collapse
Affiliation(s)
- Yu Zhao
- College of Pharmaceutical Science, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding 071002, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Baoding 071002, China
| | - Yichuan Ma
- College of Chemistry & Materials Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Baoding 071002, China
| | - Qingqing Leng
- College of Pharmaceutical Science, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding 071002, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Baoding 071002, China
| | - Qi Zhang
- College of Chemistry & Materials Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Baoding 071002, China
| | - Yuanhang Li
- College of Pharmaceutical Science, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding 071002, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Baoding 071002, China
| | - Mengmeng Ji
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Baoding 071002, China; Affiliated Hospital of Hebei University, Baoding 071000, China
| | - Hua Yang
- Affiliated Hospital of Hebei University, Baoding 071000, China
| | - Xiaoya Li
- College of Pharmaceutical Science, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding 071002, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Baoding 071002, China
| | - Guang Jia
- College of Chemistry & Materials Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Baoding 071002, China
| | - Zhenhua Li
- The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan 523059, China.
| | - Huifang Liu
- College of Pharmaceutical Science, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding 071002, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Baoding 071002, China.
| | - Jinchao Zhang
- College of Chemistry & Materials Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China; State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Baoding 071002, China.
| |
Collapse
|
4
|
Gao X, Zhang G, Wang F, Ruan W, Sun S, Zhang Q, Liu X. Emerging roles of EGFL family members in neoplastic diseases: Molecular mechanisms and targeted therapies. Biochem Pharmacol 2025; 236:116847. [PMID: 40044051 DOI: 10.1016/j.bcp.2025.116847] [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: 12/28/2024] [Revised: 02/13/2025] [Accepted: 03/03/2025] [Indexed: 03/09/2025]
Abstract
Epidermal growth factor-like proteins (EGFLs) contain more than a single EGF/EGF-like domain within their protein structure. To date, ten EGFL family members (EGFL1-10) have been characterized across diverse tissues and developmental stages under different conditions. In this review, we conclude that EGFLs are instrumental in regulating biological activities and pathological processes. Under physiological conditions, EGFLs participate in angiogenesis, neurogenesis, osteogenesis, and other processes. Under pathological conditions, EGFLs are linked with different diseases, particularly cancers. Furthermore, we highlight recent advancements in the study of EGFLs in biological conditions and cancers. In addition, the regulatory role and key underlying mechanism of EGFLs in mediating tumorigenesis are discussed. This paper also examines potential antagonists that target EGFL family members in cancer therapeutics. In summary, this comprehensive review elucidates the critical role of EGFLs in neoplastic diseases and highlights their potential as therapeutic targets.
Collapse
Affiliation(s)
- Xiaoge Gao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, PR China
| | - Guopeng Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, PR China
| | - Feitong Wang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, PR China
| | - Wenhui Ruan
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, PR China
| | - Shishuo Sun
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, PR China
| | - Qing Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, PR China
| | - Xiangye Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, PR China; Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, PR China.
| |
Collapse
|
5
|
Yan L, Su P, Sun X. Role of multi‑omics in advancing the understanding and treatment of prostate cancer (Review). Mol Med Rep 2025; 31:130. [PMID: 40116118 PMCID: PMC11938414 DOI: 10.3892/mmr.2025.13495] [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: 09/30/2024] [Accepted: 01/27/2025] [Indexed: 03/23/2025] Open
Abstract
The application of multi‑omics methodologies, encompassing genomics, transcriptomics, proteomics, metabolomics and integrative genomics, has markedly enhanced the understanding of prostate cancer (PCa). These methods have facilitated the identification of molecular pathways and biomarkers crucial for the early detection, prognostic evaluation and personalized treatment of PCa. Studies using multi‑omics technologies have elucidated how alterations in gene expression and protein interactions contribute to PCa progression and treatment resistance. Furthermore, the integration of multi‑omics data has been used in the identification of novel therapeutic targets and the development of innovative treatment modalities, such as precision medicine. The evolving landscape of multi‑omics research holds promise for not only deepening the understanding of PCa biology but also for fostering the development of more effective and tailored therapeutic interventions, ultimately improving patient outcomes. The present review aims to synthesize current findings from multi‑omics studies associated with PCa and to assess their implications for the improvement of patient management and therapeutic outcomes. The insights provided may guide future research directions and clinical practices in the fight against PCa.
Collapse
Affiliation(s)
- Li Yan
- Department of Urology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, P.R. China
| | - Pengxiao Su
- Department of Urology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, P.R. China
| | - Xiaoke Sun
- Department of Urology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, P.R. China
| |
Collapse
|
6
|
Rahman R, Selth LA. Cyclin-dependent kinases as mediators of aberrant transcription in prostate cancer. Transl Oncol 2025; 55:102378. [PMID: 40163908 PMCID: PMC11995790 DOI: 10.1016/j.tranon.2025.102378] [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: 11/24/2024] [Revised: 03/19/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025] Open
Abstract
Transcriptional control of gene expression is fundamental to all cellular processes. Conversely, transcriptional dysregulation is a hallmark of cancer. While this hallmark is a key driver of all malignancy-related process, it also represents a vulnerability that can be exploited therapeutically. Prostate cancer is a prime example of this phenomenon: it is characterised by aberrant transcription and treated with drugs that influence transcriptional pathways. Indeed, the primary oncogenic driver and therapeutic target of prostate cancer, the androgen receptor (AR), is a transcription factor. Moreover, a plethora of other transcriptional regulators, including transcriptional cyclin-dependent kinases (CDK7, CDK8 and CDK9), MYC and Bromodomain-containing protein 4 (BRD4), play prominent roles in disease progression. In this review, we focus on the roles of transcriptional CDKs in prostate cancer growth, metastasis and therapy resistance and discuss their interplay with AR, MYC and BRD4. Additionally, we explore recent advances in the therapeutic targeting of transcriptional CDKs and propose how these strategies could be effectively harnessed for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Razia Rahman
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, South Australia
| | - Luke A Selth
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, South Australia; Flinders University, Freemasons Centre for Male Health and Wellbeing, Adelaide, South Australia; Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia.
| |
Collapse
|
7
|
Suzuki K, Matsuyama H, Matsubara N, Kazama H, Ueno F, Uemura H. Current Evidence on Cabazitaxel for Prostate Cancer Therapy: A Narrative Review. Int J Urol 2025; 32:475-487. [PMID: 39996439 DOI: 10.1111/iju.70019] [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: 09/27/2024] [Revised: 01/22/2025] [Accepted: 02/11/2025] [Indexed: 02/26/2025]
Abstract
The incidence of prostate cancer (PC) has recently increased in Japan. Androgen deprivation therapy (ADT) has been a key treatment in patients with castration-sensitive PC (CSPC); however, resistance typically emerges through multiple mechanisms, leading to metastatic castration-resistant PC (mCRPC). Taxane-based therapy (i.e., docetaxel, cabazitaxel) has been standard care in patients with mCRPC. New evidence supporting the addition of androgen receptor signaling inhibitors (ARSIs, e.g., enzalutamide, abiraterone) to docetaxel and ADT for patients with metastatic CSPC (mCSPC) raises questions about the role of taxane-based therapies and their optimal sequencing, as well as how to identify patients who may benefit from taxane-based therapy. Here we review the evidence on taxane-based therapy, including cabazitaxel, in the treatment of PC, with a focus on clinical and real-world evidence from Japan. Cabazitaxel has proven effective for patients with mCRPC who have a history of ARSI and docetaxel use, and it is preferable to a second alternative ARSI, as indicated in the CARD study. The safety profile of cabazitaxel (particularly, the incidence of neutropenia) can be managed through prophylactic use of granulocyte colony-stimulating factor, as well as a lower dosage and possibly variation of the dosage interval. However, a certain dose intensity is required because neutropenia has been identified as a potential prognostic indicator for treatment effectiveness. In the ARSI era for mCSPC, evidence on mCRPC treatment sequencing is limited. A better understanding of PC biology and the collection of real-world data is essential for effective treatment and improved safety-benefit outcomes.
Collapse
Affiliation(s)
- Kazuhiro Suzuki
- Department of Urology, Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan
| | | | - Nobuaki Matsubara
- Department of Medical Oncology, National Cancer Center Hospital East, Chiba, Japan
| | | | - Fumiko Ueno
- Specialty Care, Oncology Medical, Sanofi K.K., Tokyo, Japan
| | - Hirotsugu Uemura
- Department of Urology Kindai University Faculty of Medicine, Osakasayama, Japan
| |
Collapse
|
8
|
Kumar KSP, Jyothi MN, Prashant A. CHD1 dysregulation in cancer: bridging chromatin instability, therapy resistance, and immune evasion. Mol Biol Rep 2025; 52:426. [PMID: 40278910 DOI: 10.1007/s11033-025-10536-w] [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: 02/05/2025] [Accepted: 04/22/2025] [Indexed: 04/26/2025]
Abstract
Chromodomain-Helicase-DNA-binding protein 1 (CHD1) is a central regulator of chromatin dynamics, profoundly influencing gene expression, DNA repair, and genomic stability. This review critically explores CHD1's role in cancer biology, emphasizing its complex, context-dependent functions. In prostate cancer, CHD1 acts as both a tumour suppressor and a facilitator of neuroendocrine differentiation, with its loss linked to aggressive phenotypes, resistance to androgen receptor therapies, and synthetic lethality with PTEN loss. Beyond prostate cancer, CHD1 is implicated in breast, ovarian, and hematological cancers, where it modulates chromatin accessibility, transcription regulation, and therapy resistance. Despite its promise as a biomarker and therapeutic target, CHD1 presents challenges due to its dual roles and cancer-specific effects. The review also highlights critical gaps, including the need for high-resolution studies on CHD1's interactions with immune pathways, synthetic lethality mechanisms, and chromatin remodelling in treatment resistance. Leveraging CHD1's molecular complexities could show the way for innovative diagnostic and therapeutic strategies in cancer, but its role in non-prostate cancers remains underexplored, warranting further investigation.
Collapse
Affiliation(s)
- K S Praveen Kumar
- Department of Medical Genetics, JSS Medical College and Hospital, JSS-AHER, 570015, Mysuru, India, Karnataka.
| | - M N Jyothi
- Department of Medical Genetics, JSS Medical College and Hospital, JSS-AHER, 570015, Mysuru, India, Karnataka
| | - Akila Prashant
- Department of Biochemistry, JSS Medical College and Hospital, JSS-AHER, Karnataka, 570015, Mysuru, India
| |
Collapse
|
9
|
Li C, Cheng S, Yu J, Zheng Q, Yu G, Xu M, Meng X, Zeng X, Liu K, Xu B, Luo H, Xu G. Hit to lead optimization of the 4-trifluoromethylquinoline derivatives as novel SGK1 inhibitors with potent anti-prostate cancer activity. Eur J Med Chem 2025; 287:117336. [PMID: 39908792 DOI: 10.1016/j.ejmech.2025.117336] [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: 10/22/2024] [Revised: 01/19/2025] [Accepted: 01/25/2025] [Indexed: 02/07/2025]
Abstract
Prostate cancer (PCa) remains a significant health concern for males, and serum/glucocorticoid-regulated kinase-1 (SGK1) plays a crucial role in its pathogenesis. This provides a promising target for the development of novel therapies against PCa. Herein, we reported the structural optimization of the hit compound H1, which was discovered in our previous work as an SGK1 inhibitor. Based on docking research for the active binding conformation of compound H1, a series of novel 4-trifluoromethyl quinoline derivatives were developed by replacing the 6-methoxy group in the quinoline skeleton of compound H1 with a larger aryl ring to occupy the hinge region of SGK1. Among them, compound 12f showed the strongest SGK1 inhibitory potency, with an IC50 value of 0.39 μM, representing a 7.8-fold improvement over compound H1. Molecular docking studies revealed that the 6-methoxyphenylamine moiety of compound 12f effectively extends into the hinge region of SGK1, establishing a crucial hydrogen bonding interaction with Glu183 that enhances its biological potency. In vivo, compound 12f effectively suppressed tumor growth in the PC3 xenograft model in BALB/c nude mice without inducing any observable toxicity. Moreover, mechanistic studies showed that compound 12f hindered PC3 cell migration and invasion, improved the thermal stability of SGK1 protein in PC3 cells, decreased SGK1 protein levels in tumor tissues, and effectively inhibited the phosphorylation of SGK1 and its substrates in PC3 cells in a dose- and time-dependent manner. In summary, the results of this study highlight the potential of 12f as a lead compound for further optimization in the development of new therapies against PCa targeting SGK1.
Collapse
Affiliation(s)
- Cheng Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China; Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Sha Cheng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Jia Yu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Qian Zheng
- Department of Nephrology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Gang Yu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Mei Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Xueling Meng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Xiaoping Zeng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Kun Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Bixue Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China.
| | - Heng Luo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China.
| | - Guangcan Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China.
| |
Collapse
|
10
|
Chiu CL, Zhang D, Zhao H, Wei Y, Polasko AL, Thomsen MT, Yang V, Yang KK, Hauck S, Peterson EE, Wen RM, Qiu Z, Corey E, Miao YR, Rankin EB, Peehl DM, Huang J, Giaccia AJ, Brooks JD. Targeting AXL Inhibits the Growth and Metastasis of Prostate Cancer in Bone. Clin Cancer Res 2025; 31:1346-1358. [PMID: 39879384 PMCID: PMC11961319 DOI: 10.1158/1078-0432.ccr-24-3028] [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: 09/19/2024] [Revised: 11/22/2024] [Accepted: 01/27/2025] [Indexed: 01/31/2025]
Abstract
PURPOSE After failing primary and secondary hormonal therapy, castration-resistant and neuroendocrine prostate cancer metastatic to the bone is invariably lethal, although treatment with docetaxel and carboplatin can modestly improve survival. Therefore, agents targeting biologically relevant pathways in prostate cancer and potentially synergizing with docetaxel and carboplatin in inhibiting bone metastasis growth are urgently needed. EXPERIMENTAL DESIGN Phosphorylated (activated) AXL expression in human prostate cancer bone metastases was assessed by IHC staining. We evaluated the effects of a novel soluble AXL signaling inhibitor, sAXL (batiraxcept or AVB-S6-500), on tumor growth and lung metastases in prostate cancer patient-derived xenograft models that were implanted intratibially. After injection of LuCaP cells into the tibiae, tumors were treated with batiraxcept and docetaxel or carboplatin alone or in combination, and tumor growth was monitored by serum prostate-specific antigen or bioluminescence. Tumor burden was quantified by human-specific Ku70 staining, and metastasis to the lungs was determined using qPCR. Transcriptomic profiling, Western blotting, and immunohistochemistry were performed to identify treatment-regulated gene and protein profile changes. RESULTS High AXL phosphorylation in human prostate cancer bone metastases correlated with shortened survival. Batiraxcept alone or in combination with docetaxel or carboplatin significantly suppressed intratibial tumor growth and suppressed metastasis to the lungs through multiple mechanisms, including repression of cancer stemness genes (CD44, ALDH1A1, TACSTD2, and ATXN1) and the PI3K, JAK, MAPK, and E2F1/NUSAP1 signaling pathways. CONCLUSIONS Our study provides a robust preclinical rationale and mechanisms of action for using batiraxcept as a single agent or in combination with docetaxel or carboplatin to treat lethal metastatic prostate cancer.
Collapse
Affiliation(s)
- Chun-Lung Chiu
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Dalin Zhang
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Hongjuan Zhao
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yi Wei
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Mikkel Thy Thomsen
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Clinical Medicine, Aarhus University, Denmark
| | - Vanessa Yang
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kasie Kexin Yang
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Spencer Hauck
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Eric E. Peterson
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ru M. Wen
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Zhengyuan Qiu
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, Washington, USA
| | - Yu Rebecca Miao
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Erinn B. Rankin
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Donna M. Peehl
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Jiaoti Huang
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Amato J. Giaccia
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Oncology, University of Oxford, Oxford, UK
| | - James D. Brooks
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Research Institute, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
11
|
Jiang H, Chen H, Wang Y, Xu H, Chen H. Synthesis, bioactivity, and molecular docking studies: novel arylpiperazine derivatives as potential new-resistant AR antagonists. Front Chem 2025; 13:1557275. [PMID: 40224223 PMCID: PMC11985751 DOI: 10.3389/fchem.2025.1557275] [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: 01/08/2025] [Accepted: 03/17/2025] [Indexed: 04/15/2025] Open
Abstract
The majority of patients with androgen-dependent prostate cancer (PCa) develop resistance to hormone therapy after approximately 18-24 months of androgen deprivation therapy treatment. During this process, PCa cells progressively lose their sensitivity to androgens and evolve into castration-resistant prostate cancer leading to uncontrolled tumor growth and ultimately the failure of endocrine therapy. To develop potential anti-prostate cancer agents, in this study, we identified a novel ether-type arylpiperazine derivative as a potent androgen receptor (AR) antagonist, uncovering a series of effective antiproliferative compounds. The derivatives (7, 11, 17, 19, 20, 21, 22, 23, and 24) demonstrated strong cytotoxicity against cancer cells, with 17, 19, 20, and 23 showing significant androgen receptor antagonistic activity (Inhibition% >60) and robust AR binding affinities. The structure-activity relationship (SAR) of these developed derivatives was discussed based on data. Docking study suggested that the compound 19 mainly bind to AR ligand binding pocket site through Van der Waals' force interactions. This research presents a promising lead compound for developing anticancer agents targeting prostate cancer therapy.
Collapse
Affiliation(s)
- Hua Jiang
- Department of Urology, The Fifth Affiliated Hospital of Zunyi Medical University (Zhuhai Sixth People’s Hospital), Zhuhai, China
| | - Haowei Chen
- Luoyang Key Laboratory of Organic Functional Molecules, College of Food and Drug, Luoyang Normal University, Luoyang, China
| | - Ya Wang
- Luoyang Key Laboratory of Organic Functional Molecules, College of Food and Drug, Luoyang Normal University, Luoyang, China
| | - Huaxin Xu
- Luoyang Key Laboratory of Organic Functional Molecules, College of Food and Drug, Luoyang Normal University, Luoyang, China
| | - Hong Chen
- Luoyang Key Laboratory of Organic Functional Molecules, College of Food and Drug, Luoyang Normal University, Luoyang, China
| |
Collapse
|
12
|
Fan H, Liu J, Hu X, Cai J, Su B, Jiang J. The critical role of X-linked inhibitor of apoptosis protein (XIAP) in tumor development. Apoptosis 2025:10.1007/s10495-025-02101-4. [PMID: 40146486 DOI: 10.1007/s10495-025-02101-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2025] [Indexed: 03/28/2025]
Abstract
X-linked inhibitor of apoptosis protein (XIAP) is the most potent endogenous member of the inhibitor of apoptosis protein family. XIAP exerts its anti-apoptotic effects by inhibiting both the death receptor pathway and mitochondrial pathway of apoptosis through various mechanisms such as directly binding to caspases, activating the nuclear factor kappa B (NF-κB) pathway, and other signaling pathways. These processes are closely related to tumor development and progression, making XIAP a therapeutic target for various types of cancer. This article will first review the structural characteristics and biological functions of XIAP, followed by its effects on tumors and an overview of XIAP-targeted inhibitors.
Collapse
Affiliation(s)
- Hui Fan
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jiyuan Liu
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xiangyan Hu
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jiye Cai
- Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Bo Su
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jinhuan Jiang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
13
|
Tao M, Zhang P, Yang W, Wang Y, Chen J, Shi X, Dai E, Yan W, Fu Y. Global, regional, and national burden of disease attributable to diet low in milk, 1990-2021: an updated analysis of the Global Burden of Disease study 2021. J Dairy Sci 2025:S0022-0302(25)00159-6. [PMID: 40139368 DOI: 10.3168/jds.2024-26131] [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: 12/09/2024] [Accepted: 02/07/2025] [Indexed: 03/29/2025]
Abstract
Diet low in milk has been linked to various chronic diseases and cancer. To better understand the global health impact of diet low in milk, this study analyzed the latest data from the Global Burden of Disease (GBD) study 2021. Our findings revealed that the number of deaths and DALYs of colon and rectum cancer (CRC) attributable to diet low in milk increased from 1990 to 2021. The ASMR and ASDR for CRC due to low milk intake declined slightly over this period, with a faster decline observed among females. Predictive analysis shows similar tendency, even higher ASMR and ASDR in CRC associated with diet low in milk in males than in females by 2050. In contrast, the rate of prostate cancer attributable to diet low in milk remained relatively stable. Regionally, Southern Latin America exhibited the highest ASMR and ASDR for CRC associated with diet low in milk in 2021, while Western Sub-Saharan Africa had the most significant changes in these rates for prostate cancer. The largest decrease in ASMR and ASDR of CRC due to diet low in milk was observed in the high SDI region. In addition, the low SDI region exhibited the highest change rates for prostate cancer attributable to diet low in milk. In summary, our study provides valuable insights into trends for the global burden of CRC and prostate cancer attributable to diet low in milk, emphasizing the importance of considering gender, age, regional and national differences in dietary habits and disease risk when formulating public health policies and dietary recommendations.
Collapse
Affiliation(s)
- Meihui Tao
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Po Zhang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weifeng Yang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanping Wang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junfa Chen
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiu Shi
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Erfang Dai
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Yan
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yu Fu
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
14
|
Xie Y, Chen G, Wu W, Wen X, Lai M, Che L, Ran J. A bioinformatics approach combined with experimental validation analyzes the efficacy of azithromycin in treating SARS-CoV-2 infection in patients with IPF and COPD These authors contributed equally: Yining Xie, Guangshu Chen, and Weiling Wu. Sci Rep 2025; 15:10009. [PMID: 40122903 PMCID: PMC11930991 DOI: 10.1038/s41598-025-94801-9] [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: 12/05/2024] [Accepted: 03/17/2025] [Indexed: 03/25/2025] Open
Abstract
The swift transmission rate and unfavorable prognosis associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have prompted the pursuit of more effective therapeutic interventions. Azithromycin (AZM) has garnered significant attention for its distinctive pharmacological mechanisms in the treatment of SARS-CoV-2. This study aims to elucidate the biological rationale for employing AZM in patients with chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF) who are infected with SARS-CoV-2. Genetic data about COVID-19, COPD, and IPF were independently obtained from the GeneCards database. And 40 drug targets about AZM were retrieved from the STITCH database. The analysis revealed that 311 DEGs were common among COPD, IPF, and COVID-19, and we further found eight genes that interacted with AZM targets. We conducted an analysis of hub genes and their corresponding signaling pathways in these patient cohorts. Additionally, we explored the inhibitory effects of AZM on these hub genes. AZM demonstrated a significant inhibitory effect on eight key genes, except for AR and IL-17 A. These findings suggest that AZM may serve as a promising therapeutic agent for patients with COPD and IPF and SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Yining Xie
- Department of Endocrinology and Metabolism, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
- Department of Endocrinology and Metabolism, The First People's Hospital of Zhaoqing, Zhaoqing, 526060, China
| | - Guangshu Chen
- Department of Endocrinology and Metabolism, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Weiling Wu
- Department of Endocrinology and Metabolism, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Xueman Wen
- Department of Endocrinology and Metabolism, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Meizheng Lai
- Department of Endocrinology and Metabolism, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Li Che
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China.
| | - Jianmin Ran
- Department of Endocrinology and Metabolism, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China.
| |
Collapse
|
15
|
Kwon WA, Joung JY. Immunotherapy in Prostate Cancer: From a "Cold" Tumor to a "Hot" Prospect. Cancers (Basel) 2025; 17:1064. [PMID: 40227610 PMCID: PMC11987915 DOI: 10.3390/cancers17071064] [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: 02/17/2025] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Immunotherapy has shown limited efficacy in prostate cancer, largely due to low tumor immunogenicity, sparse tumor-infiltrating lymphocytes, and a suppressive microenvironment. Recent therapeutic strategies aim to boost immune responses and counteract immunosuppressive factors through interventions such as immune checkpoint inhibitors, immunogenic cell death-inducing therapies, and the targeted blockade of pathways like that of transforming growth factor-β. Vaccine-based approaches, potent immune adjuvants, and engineered chimeric antigen receptor (CAR) T cells are also being investigated to overcome local immune inhibitory signals. Advancements in imaging, multi-omic profiling, and liquid biopsies offer promising avenues for real-time monitoring, better patient selection, and precision treatment. This review provides an overview of the key immunosuppressive features of prostate cancer, current immunotherapeutic modalities, and emerging strategies to transform "cold" tumors into more responsive "hot" targets. By integrating these approaches, we may achieve more durable clinical benefits for patients with advanced or metastatic prostate cancer.
Collapse
Affiliation(s)
- Whi-An Kwon
- Department of Urology, Hanyang University College of Medicine, Myongji Hospital, Goyang 10475, Republic of Korea
| | - Jae Young Joung
- Department of Urology, Urological Cancer Center, National Cancer Center, Goyang 10408, Republic of Korea
| |
Collapse
|
16
|
Bugoye FC, Torrorey-Sawe R, Biegon R, Dharsee N, Mafumiko F, Kibona H, Aboud S, Patel K, Mining S. Exploring therapeutic applications of PTEN, TMPRSS2:ERG fusion, and tumour molecular subtypes in prostate cancer management. Front Oncol 2025; 15:1521204. [PMID: 40165885 PMCID: PMC11956161 DOI: 10.3389/fonc.2025.1521204] [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: 11/01/2024] [Accepted: 02/21/2025] [Indexed: 04/02/2025] Open
Abstract
Background Prostate cancer is defined by the suppression of genes that suppress tumours and the activation of proto-oncogenes. These are the hallmarks of prostate cancer, and they have been linked to numerous genomic variations, which lead to unfavourable treatment outcomes. Prostate cancer can be categorised into various risk groups of tumour molecular subtypes grounded in the idea of genomic structural variations connected to TMPRSS2:ERG fusion and loss of PTEN. Research suggests that certain genomic alterations may be more prevalent or exhibit different patterns in prostate cancer tumours across populations. Studies have reported a higher frequency of PTEN loss and TMPRSS2:ERG fusion in prostate tumours of Black/African American men, which may contribute to the more aggressive nature of the disease in this population. Thus, therapeutically important information can be obtained from these structural variations, including correlations with poor prognosis and disease severity. Methods Peer-reviewed articles from 1998 to 2024 were sourced from PubMed and Google Scholar. During the review process, the following search terms were employed: "Tumour suppressor genes OR variations OR alterations OR oncogenes OR diagnostics OR ethnicity OR biomarkers OR prostate cancer genomics OR prostate cancer structural variations OR tumour and molecular subtypes OR therapeutic implications OR immunotherapy OR immunogenetics." Results There was a total of 13,012 results for our search query: 5,903 publications from Google Scholar with the patent and citation unchecked filer options, and 7127 articles from PubMed with the abstract, free full text, and full-text options selected. Unpublished works were not involved. Except for four articles published between 1998 and 1999, all other selected articles published in 2000 and later were considered. However, papers with irrelevant information or redundant or duplicate content were not chosen for this review. Thus, 134 met the inclusion criteria and were ultimately retained for this review. Conclusion This review extracted 134 relevant articles about genomic structure variations in prostate cancer. Our findings demonstrate the importance of PTEN and TMPRSS2:ERG fusion and tumour molecular subtyping in prostate cancer precision medicine.
Collapse
Affiliation(s)
- Fidelis Charles Bugoye
- Directorate of Forensic Science and DNA Services, Government Chemist Laboratory Authority, Dar es Salaam, Tanzania
- Department of Pathology, Moi University, Moi Teaching and Referral Hospital, Eldoret, Kenya
| | - Rispah Torrorey-Sawe
- Department of Pathology, Moi University, Moi Teaching and Referral Hospital, Eldoret, Kenya
| | - Richard Biegon
- Department of Pathology, Moi University, Moi Teaching and Referral Hospital, Eldoret, Kenya
| | - Nazima Dharsee
- Clinical Research, Training and Consultancy Unit, Ocean Road Cancer Institute, Dar es Salaam, Tanzania
| | - Fidelice Mafumiko
- Directorate of Forensic Science and DNA Services, Government Chemist Laboratory Authority, Dar es Salaam, Tanzania
| | - Herry Kibona
- Department of Urology, Muhimbili National Hospital, Dar es Salaam, Tanzania
| | - Said Aboud
- Head Office, National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Kirtika Patel
- Department of Pathology, Moi University, Moi Teaching and Referral Hospital, Eldoret, Kenya
| | - Simeon Mining
- Department of Pathology, Moi University, Moi Teaching and Referral Hospital, Eldoret, Kenya
| |
Collapse
|
17
|
Xu S, Shi H, Liu Y, Lin J, Wu X, Lu R, Fan Y, Tan W. Identification of biomarkers associated with pathological tumor staging and their utility in the diagnosis and prognosis of prostate cancer. Lab Med 2025; 56:118-128. [PMID: 39141479 DOI: 10.1093/labmed/lmae059] [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] [Indexed: 08/16/2024] Open
Abstract
OBJECTIVE Pathological tumor (pT) staging plays a crucial role in prostate cancer (PCa) diagnosis. This study aimed to identify pT stage-associated biomarkers and explored their utility in PCa prognosis. METHODS GSE69223 was used to identify potential targets differentially expressed between level 2 of pT staging (pT2) and level 3 of pT staging (pT3). Quantitative reverse transcriptase-polymerase chain reaction and immunohistochemistry were performed on tissues from patients with PCa to screen the pT stage-associated targets and to explore the prognostic value of these targets in PCa. RESULTS CENPI and SLC38A11 were most significantly upregulated, whereas ANO6 and KANK2 were mostly decreased in pT3 tumors compared with pT2 staging. ANO6 levels were negatively associated with preoperative prostate-specific antigen (PSA) levels, lymph node staging (N staging), Gleason score, and overall survival (OS); CENPI was positively associated with preoperative PSA levels, N staging, and OS, but was not associated with the Gleason score; SLC38A11 and KANK2 were not associated with OS. ANO6 and KANK2 were correlated with neutrophil markers, whereas CENPI was correlated with macrophage M2 types. CONCLUSION We identified 4 reliable PCa biomarkers associated with pT staging that would be valuable for diagnosing and determining PCa prognosis.
Collapse
Affiliation(s)
- Shiquan Xu
- Department of Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - He Shi
- Orthopedics and Sports Medicine Center, Qingdao Municipal Hospital, Qingdao, China
| | - Yiran Liu
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
| | - Jing Lin
- Department of Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xia Wu
- Department of Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Ruichun Lu
- Department of Cadre Healthcare/Geriatrics, Qingdao Municipal Hospital, Qingdao, China
| | - Yu Fan
- Department of Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Weiqiang Tan
- Department of Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
18
|
Yi J, Du J, Chen X, Nie RC, Hu GS, Wang L, Zhang YY, Chen S, Wen XS, Luo DX, He H, Liu W. A circRNA-mRNA pairing mechanism regulates tumor growth and endocrine therapy resistance in ER-positive breast cancer. Proc Natl Acad Sci U S A 2025; 122:e2420383122. [PMID: 40233410 PMCID: PMC11874584 DOI: 10.1073/pnas.2420383122] [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: 10/04/2024] [Accepted: 01/13/2025] [Indexed: 04/17/2025] Open
Abstract
The molecular mechanisms underlying estrogen receptor (ER)-positive breast carcinogenesis and drug resistance remain incompletely understood. Elevated expression of CCND1 is linked to enhanced invasiveness, poorer prognosis, and resistance to drug therapies in ER-positive breast cancer. In this study, we identify a highly expressed circular RNA (circRNA) derived from FOXK2, called circFOXK2, which plays a key role in stabilizing CCND1 mRNA, thereby promoting cell cycle progression, cell growth, and endocrine therapy resistance in ER-positive breast cancer cells. Mechanistically, circFOXK2 binds directly to CCND1 mRNA via RNA-RNA pairing and recruits the RNA-binding protein ELAVL1/HuR, stabilizing the CCND1 mRNA and enhancing CCND1 protein levels. This results in activation of the CCND1-CDK4/6-p-RB-E2F signaling axis, driving the transcription of downstream E2F target genes and facilitating the G1/S transition during cell cycle progression. Notably, targeting circFOXK2 with antisense oligonucleotide (ASO-circFOXK2) suppresses ER-positive breast cancer cell growth both in vitro and in vivo. Moreover, combination therapy with ASO-circFOXK2 and tamoxifen exhibits synergistic effects and restores tamoxifen sensitivity in tamoxifen-resistant cells. Clinically, high circFOXK2 expression is positively correlated with CCND1 levels in both ER-positive breast cancer cell lines and patient tumor tissues. Overall, our findings reveal the critical role of circFOXK2 in stabilizing the oncogene CCND1 and promoting cancer progression, positioning circFOXK2 as a potential therapeutic target for ER-positive breast cancer in clinical settings.
Collapse
MESH Headings
- Humans
- Breast Neoplasms/genetics
- Breast Neoplasms/drug therapy
- Breast Neoplasms/pathology
- Breast Neoplasms/metabolism
- Female
- Drug Resistance, Neoplasm/genetics
- RNA, Circular/genetics
- RNA, Circular/metabolism
- Cyclin D1/genetics
- Cyclin D1/metabolism
- Animals
- Mice
- Cell Line, Tumor
- Cell Proliferation/genetics
- Cell Proliferation/drug effects
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Estrogen/metabolism
- Receptors, Estrogen/genetics
- Gene Expression Regulation, Neoplastic
- Antineoplastic Agents, Hormonal/pharmacology
- ELAV-Like Protein 1/metabolism
- ELAV-Like Protein 1/genetics
- Tamoxifen/pharmacology
- Mice, Nude
- MCF-7 Cells
Collapse
Affiliation(s)
- Jia Yi
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Yu-Yue Pathology Scientific Research Center, Chongqing400039, China
- Jinfeng Laboratory, Chongqing401329, China
| | - Jiao Du
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
| | - Xue Chen
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
| | - Rui-chao Nie
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen361102, Fujian, China
| | - Guo-sheng Hu
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
| | - Lei Wang
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
| | - Yue-ying Zhang
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
| | - Shang Chen
- Laboratory Medicine Centre, Shenzhen Nanshan People’s Hospital, Shenzhen518052, Guangdong, China
| | - Xiao-sha Wen
- Laboratory Medicine Centre, Shenzhen Nanshan People’s Hospital, Shenzhen518052, Guangdong, China
| | - Di-xian Luo
- The Third Affiliated Hospital (Luohu Hospital), Shenzhen University, Shenzhen518000, Guangdong, China
| | - Hua He
- Department of Neurosurgery, Third Affiliated Hospital, Naval Medical University, Shanghai200438, China
| | - Wen Liu
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen361102, Fujian, China
| |
Collapse
|
19
|
Siskin M, Economides MP, Wise DR. Cyclin-Dependent Kinase Inhibition in Prostate Cancer: Past, Present, and Future. Cancers (Basel) 2025; 17:774. [PMID: 40075623 PMCID: PMC11898528 DOI: 10.3390/cancers17050774] [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: 01/15/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Despite significant progress, prostate cancer remains a leading cause of death. Cyclin-dependent kinase (CDK) 4/6 inhibitors, which are already approved for the treatment of hormone receptor-positive breast cancer, are undergoing extensive testing as monotherapy and in various combinations as a potentially valuable treatment modality in prostate cancer patients. Thus far, a limited number of these studies have published results, which have been largely disappointing. AREAS COVERED In this review, we describe the biologic rationale for the use of CDK4/6 inhibitors in prostate cancer, the existing clinical data describing their use in prostate cancer, and ongoing clinical trials of CDK4/6 inhibitors as monotherapy and in combination for the treatment of prostate cancer. In particular, we focus on possible resistance mechanisms that may be particularly relevant in prostate cancer patients, leading to de novo and acquired resistance, and we highlight novel strategies that can overcome this resistance. CONCLUSIONS Current clinical trials are actively working to (1) refine the role of CDK4/6 inhibitors in prostate cancer patients; (2) develop new inhibitors of other cell-cycle targets, such as CDK2 and CDK7; and (3) explore novel combination therapies with inhibitors of other relevant pathways, such as PI3K or MAPK. Further genomic subtyping of advanced prostate cancer will likely shed light on the subsets of patients most likely to benefit from cell-cycle-targeted agents.
Collapse
Affiliation(s)
| | | | - David R. Wise
- Genitourinary Medical Oncology Service, Perlmutter Cancer Center, NYU Langone Heath Center, New York, NY 10016, USA; (M.S.); (M.P.E.)
| |
Collapse
|
20
|
Khorasanchi A, Hong F, Yang Y, Singer EA, Wang P, Li M, Zheng L, Monk P, Mortazavi A, Meng L. Overcoming drug resistance in castrate-resistant prostate cancer: current mechanisms and emerging therapeutic approaches. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:9. [PMID: 40051495 PMCID: PMC11883235 DOI: 10.20517/cdr.2024.173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/24/2025] [Accepted: 02/07/2025] [Indexed: 03/09/2025]
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) is driven by a complex network of resistance mechanisms against standard-of-care therapies, resulting in poor long-term outcomes. This review offers a uniquely comprehensive and integrative perspective on these resistance pathways, systematically examining both androgen receptor (AR)-dependent factors (including AR overexpression, point mutations, glucocorticoid receptor signaling, splice variants, post-translational modifications, altered coregulators, and intratumoral hormone biosynthesis) and AR-independent pathways (such as neuroendocrine differentiation, lineage plasticity, and alternative growth factor signaling). We also highlight resistance mechanisms influencing immunotherapy, chemotherapy, radiopharmaceutical therapy and targeted therapy. By synthesizing emerging insights across these domains, this review not only clarifies the underlying biology of mCRPC resistance but also identifies key leverage points for more effective interventions. Building on this foundation, we propose a forward-looking framework for overcoming mCRPC drug resistance, emphasizing the importance of biomarker-guided patient selection, combination strategies that simultaneously target multiple resistance mechanisms, and novel therapies under investigation. These recommendations are intended to guide future clinical trial designs and research priorities that move beyond incremental improvements. Ultimately, this comprehensive synthesis aims to serve as a resource for clinicians and researchers to accelerate the development of durable, precision-based treatment strategies in mCRPC.
Collapse
Affiliation(s)
- Adam Khorasanchi
- Division of Hospital Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Feng Hong
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH 43210, USA
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Yuanquan Yang
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH 43210, USA
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Eric A. Singer
- Division of Urologic Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Peng Wang
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Mingjia Li
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Linghua Zheng
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH 43210, USA
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Paul Monk
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Amir Mortazavi
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Lingbin Meng
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| |
Collapse
|
21
|
Sun X, Li Z, Wang L, Yan H, Li X, Wang N, Dong Z, Xu W. Functional Study of Four Histone Genes Involved in the Spermatogenesis of Cynoglossus semilaevis. Animals (Basel) 2025; 15:593. [PMID: 40003074 PMCID: PMC11851421 DOI: 10.3390/ani15040593] [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: 01/06/2025] [Revised: 01/29/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Chinese tongue sole (Cynoglossus semilaevis) is an important mariculture fish in China, and female individuals present a growth advantage. However, genetic females (ZW) can sex reverse to phenotypic males, designated pseudomales. The pseudomale shows abnormal spermatogenesis and produces only Z sperm. Histone is pivotal in spermatogenesis, and post-translational modification could regulate its function. A comparison of testis phosphorylated and ubiquitinated proteins revealed 8 and 12 differentially phosphorylated and ubiquitinated histones in the testes of male and pseudomale Chinese tongue soles, respectively, but there was no difference in the translation level of these proteins. We selected four histone genes, h1.1-like, h1.2-like, h3, and h3.3-like, for further analysis. The expression levels of the h1.1-like, h3, and h3.3-like genes reached their highest levels at 2 years post-hatching (yph), and the expression level of h1.2-like reached its highest level at 1.5 years post-hatching (1.5 yph), indicating that its role began during the late stage of gonadal development. Promoter activity verification revealed that the promoters of the h1.1-like, h1.2-like, h3, and h3.3-like genes were located approximately upstream 2000 bp and six histone-related transcription factor sites were predicted. YY1A, YY1B, C-JUN, and JUNB may have negative regulatory effects on h1.1-like, h1.2-like, h3, and h3.3-like; AR and ETS-2 may have positive regulatory effects on h3 and h3.3-like. The ISH results revealed that h1.1-like, h1.2-like, h3, and h3.3-like mRNAs were located mainly in the sperm cells in the testes and the oocytes at various stages in the ovaries. After siRNA knockdown, the expression of dmrt1 in testis cell lines and the expression of tesk1 and neurl3 in males was downregulated, suggesting that the h1.1-like, h1.2-like, h3, and h3.3-like genes may have a negative regulatory role in spermatogenesis. The regulatory role in female fish remains to be explored. Mass spectrometry analysis revealed that histones have an important role in chromosome remodeling. These results provide a genetic basis for the molecular mechanism of gonadal development and spermatogenesis in Chinese tongue sole.
Collapse
Affiliation(s)
- Xuexue Sun
- Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China; (X.S.); (L.W.); (H.Y.)
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Laboratory for Marine Fisheries Science and Food Production Processes, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao Marine Science and Technology Center, Qingdao 266071, China; (Z.L.); (X.L.); (N.W.)
| | - Zhijie Li
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Laboratory for Marine Fisheries Science and Food Production Processes, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao Marine Science and Technology Center, Qingdao 266071, China; (Z.L.); (X.L.); (N.W.)
| | - Lijun Wang
- Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China; (X.S.); (L.W.); (H.Y.)
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Laboratory for Marine Fisheries Science and Food Production Processes, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao Marine Science and Technology Center, Qingdao 266071, China; (Z.L.); (X.L.); (N.W.)
| | - Haipeng Yan
- Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China; (X.S.); (L.W.); (H.Y.)
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Laboratory for Marine Fisheries Science and Food Production Processes, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao Marine Science and Technology Center, Qingdao 266071, China; (Z.L.); (X.L.); (N.W.)
| | - Xihong Li
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Laboratory for Marine Fisheries Science and Food Production Processes, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao Marine Science and Technology Center, Qingdao 266071, China; (Z.L.); (X.L.); (N.W.)
| | - Na Wang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Laboratory for Marine Fisheries Science and Food Production Processes, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao Marine Science and Technology Center, Qingdao 266071, China; (Z.L.); (X.L.); (N.W.)
| | - Zhongdian Dong
- Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China; (X.S.); (L.W.); (H.Y.)
| | - Wenteng Xu
- Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China; (X.S.); (L.W.); (H.Y.)
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Laboratory for Marine Fisheries Science and Food Production Processes, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao Marine Science and Technology Center, Qingdao 266071, China; (Z.L.); (X.L.); (N.W.)
| |
Collapse
|
22
|
Ajayi AF, Oyovwi MO, Akano OP, Akanbi GB, Adisa FB. Molecular pathways in reproductive cancers: a focus on prostate and ovarian cancer. Cancer Cell Int 2025; 25:33. [PMID: 39901204 PMCID: PMC11792371 DOI: 10.1186/s12935-025-03658-5] [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: 08/12/2024] [Accepted: 01/21/2025] [Indexed: 02/05/2025] Open
Abstract
Reproductive cancers, including prostate and ovarian cancer, are highly prevalent worldwide and pose significant health challenges. The molecular underpinnings of these cancers are complex and involve dysregulation of various cellular pathways. Understanding these pathways is crucial for developing effective therapeutic strategies. This review aims to provide an overview of the molecular pathways implicated in prostate and ovarian cancers, highlighting key genetic alterations, signaling cascades, and epigenetic modifications. A comprehensive literature search was conducted using databases such as PubMed, Web of Science, and Google Scholar. Articles focusing on molecular pathways in prostate and ovarian cancer were reviewed and analyzed. In prostate cancer, recurrent mutations in genes like AR, TP53, and PTEN drive tumor growth and progression. Androgen signaling plays a significant role, with alterations in the AR pathway contributing to resistance to antiandrogen therapies. In ovarian cancer, high-grade serous carcinomas are characterized by mutations in TP53, BRCA1/2, and homologous recombination repair genes. PI3K and MAPK pathways are frequently activated, promoting cell proliferation and survival. Epigenetic alterations, including DNA methylation and histone modifications, are also prevalent in both cancer types. The molecular pathways involved in prostate and ovarian cancer are diverse and complex. Targeting these pathways with precision medicine approaches holds promise for improving patient outcomes. Further research is needed to elucidate the mechanisms of resistance and identify novel therapeutic vulnerabilities.
Collapse
Affiliation(s)
- Ayodeji Folorunsho Ajayi
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo, Nigeria
- Anchor Biomed Research Institute, Ogbomoso, Oyo, Nigeria
- Department of Physiology, Adeleke University, Ede, Osun, Nigeria
| | | | - Oyedayo Phillips Akano
- Department of Physiology, School of Basic Medical Sciences, Babcock University, Ilishan Remo, Ogun, Nigeria
| | - Grace Bosede Akanbi
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo, Nigeria
| | - Florence Bukola Adisa
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo, Nigeria
| |
Collapse
|
23
|
Wei J, Wang J, Guan W, Li J, Pu T, Corey E, Lin TP, Gao AC, Wu BJ. PlexinD1 is a driver and a therapeutic target in advanced prostate cancer. EMBO Mol Med 2025; 17:336-364. [PMID: 39748059 PMCID: PMC11822115 DOI: 10.1038/s44321-024-00186-z] [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: 04/09/2024] [Revised: 12/06/2024] [Accepted: 12/11/2024] [Indexed: 01/04/2025] Open
Abstract
Aggressive prostate cancer (PCa) variants associated with androgen receptor signaling inhibitor (ARSI) resistance and metastasis remain poorly understood. Here, we identify the axon guidance semaphorin receptor PlexinD1 as a crucial driver of cancer aggressiveness in metastatic castration-resistant prostate cancer (CRPC). High PlexinD1 expression in human PCa is correlated with adverse clinical outcomes. PlexinD1 critically maintains CRPC aggressive behaviors in vitro and in vivo, and confers stemness and cellular plasticity to promote multilineage differentiation including a neuroendocrine-like phenotype for ARSI resistance. Mechanistically, PlexinD1 is upregulated upon relief of AR-mediated transcriptional repression of PlexinD1 under ARSI treatment, and subsdquently transactivates ErbB3 and cMet via direct interaction, which triggers the ERK/AKT pathways to induce noncanonical Gli1-dictated Hedgehog signaling, facilitating the growth and plasticity of PCa cells. Blockade of PlexinD1 by the protein inhibitor D1SP restricted CRPC growth in multiple preclinical models. Collectively, these findings characterize PlexinD1's contribution to PCa progression and offer a potential PlexinD1-targeted therapy for advanced PCa.
Collapse
Affiliation(s)
- Jing Wei
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA
| | - Jing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
| | - Wen Guan
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA
| | - Jingjing Li
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA
- Engineering Research Center of Cell & Therapeutic Antibody, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Tianjie Pu
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA, 98195, USA
| | - Tzu-Ping Lin
- Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan, 11217, Republic of China
- Department of Urology, School of Medicine and Shu-Tien Urological Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan, 11221, Republic of China
| | - Allen C Gao
- Department of Urologic Surgery, University of California, Davis, Sacramento, CA, 95817, USA
| | - Boyang Jason Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA.
| |
Collapse
|
24
|
Modanwal S, Mishra A, Mishra N. Exploration of rutin derivatives as potential inhibitors of prostate cancer signaling pathways: A comprehensive in-silico study. Biochem Biophys Res Commun 2025; 746:151279. [PMID: 39754971 DOI: 10.1016/j.bbrc.2024.151279] [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: 09/07/2024] [Revised: 12/25/2024] [Accepted: 12/31/2024] [Indexed: 01/06/2025]
Abstract
Prostate cancer is a widespread health issue that affects men worldwide. It is one of the most common forms of cancer, and its development is influenced by a combination of hereditary, epigenetic, environmental, age, and lifestyle factors. Given that it is the second most common cause of cancer-related deaths in men, it is crucial to comprehend its complex facets. Present research especially targets the 3-kinase/protein kinase B, Epidermal Growth Factor Receptor, and extracellular signal-related kinase pathways, which are known to be significantly involved in prostate cancer progression. Here, Rutin derivatives were screened against selected prostate cancer targets. Molecular docking was performed to identify favorable interactions and the most promising compound. Further, Density functional theory, pharmacokinetics, Molecular dynamics simulation, principal component analysis, free energy landscape analysis, and Molecular Mechanics Poisson-Boltzmann Surface Area provided additional insights into selecting the best drug candidate. Among all the selected rutin derivatives, RU4b1 has potent inhibitory action. We also performed predictive analysis to identify the distinct metabolic sites within the structure of RU4b1. RU4b1 also exhibits drug-like properties and potent antioxidant activity. The findings were also compared with standard drugs and reference molecules of the respective proteins, and it is noteworthy that RU4b1 exhibited superior action compared to the standard drugs and reference molecules. This study aims to contribute valuable insights into developing targeted therapies for prostate cancer, emphasizing the potential of rutin derivatives as effective anti-cancer agents.
Collapse
Affiliation(s)
- Shristi Modanwal
- Department of Applied Sciences, Indian Institute of Information of Technology Allahabad, Prayagraj, Uttar Pradesh, 211012, India
| | - Ashutosh Mishra
- Department of Applied Sciences, Indian Institute of Information of Technology Allahabad, Prayagraj, Uttar Pradesh, 211012, India
| | - Nidhi Mishra
- Department of Applied Sciences, Indian Institute of Information of Technology Allahabad, Prayagraj, Uttar Pradesh, 211012, India.
| |
Collapse
|
25
|
Fang J, Alhaskawi A, Dong Y, Cheng C, Xu Z, Tian J, Abdalbary SA, Lu H. Advancements in molecular imaging probes for precision diagnosis and treatment of prostate cancer. J Zhejiang Univ Sci B 2025; 26:124-144. [PMID: 40015933 PMCID: PMC11867783 DOI: 10.1631/jzus.b2300614] [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: 08/29/2023] [Accepted: 11/30/2023] [Indexed: 01/19/2025]
Abstract
Prostate cancer is the second most common cancer in men, accounting for 14.1% of new cancer cases in 2020. The aggressiveness of prostate cancer is highly variable, depending on its grade and stage at the time of diagnosis. Despite recent advances in prostate cancer treatment, some patients still experience recurrence or even progression after undergoing radical treatment. Accurate initial staging and monitoring for recurrence determine patient management, which in turn affect patient prognosis and survival. Classical imaging has limitations in the diagnosis and treatment of prostate cancer, but the use of novel molecular probes has improved the detection rate, specificity, and accuracy of prostate cancer detection. Molecular probe-based imaging modalities allow the visualization and quantitative measurement of biological processes at the molecular and cellular levels in living systems. An increased understanding of tumor biology of prostate cancer and the discovery of new tumor biomarkers have allowed the exploration of additional molecular probe targets. The development of novel ligands and advances in nano-based delivery technologies have accelerated the research and development of molecular probes. Here, we summarize the use of molecular probes in positron emission tomography (PET), single-photon emission computed tomography (SPECT), magnetic resonance imaging (MRI), optical imaging, and ultrasound imaging, and provide a brief overview of important target molecules in prostate cancer.
Collapse
Affiliation(s)
- Jiajie Fang
- Department of Urology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Ahmad Alhaskawi
- Department of Orthopedics, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Yanzhao Dong
- Department of Orthopedics, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Cheng Cheng
- Department of Urology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310024, China
| | - Zhijie Xu
- Department of Urology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310024, China
| | - Junjie Tian
- Department of Urology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310024, China
| | - Sahar Ahmed Abdalbary
- Department of Orthopedic Physical Therapy, Faculty of Physical Therapy, Nahda University, Beni Suef 62511, Egypt
- Biomechanics and Microsurgery Labs, Nahda University, Beni Suef 62511, Egypt
| | - Hui Lu
- Department of Orthopedics, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China.
- Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
26
|
Ma Z, Zhao X, Lin Y, Zhang H, Wu L, Tao Y, Shi H, Li S. From past to present: tracing the trends of diabetes drug trials in mainland China. Front Endocrinol (Lausanne) 2025; 15:1427148. [PMID: 39866740 PMCID: PMC11757101 DOI: 10.3389/fendo.2024.1427148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 12/20/2024] [Indexed: 01/28/2025] Open
Abstract
Background This study aimed to analyze the changing trend of diabetes drugs clinical trials in China during 2013-2023, and provided a reference for the research and development of diabetes drugs. Methods Diabetes drug clinical trial data were obtained from the registration and information disclosure platform of the National Medical Products Administration (NMPA) between January 1, 2013, and December 31, 2023. Trends of clinical trials on diabetes drugs were systematically analyzed in terms of characteristics, trial design, time trends, drug type, and indications. Results From 2013 to 2023, a total of 1,256 diabetes drugs clinical trials have been registered on the NMPA platform, of which 1056 were chemical drugs and 184 were biological products. The indications are mainly type 2 diabetes mellitus (n=1237, 98.49%). Among them, 838 clinical trials have been completed, 379 were proceeding, and 39 have been terminated or suspended. There were 42 international multi-center clinical trials, and the remaining 1034 clinical trials were domestic. Bioequivalence trials were 691, accounting for 55.02%, followed by 340 phase I clinical trials and 169 phase III clinical trials. The leading units were mostly distributed in eastern China. The proportion of clinical trial sponsorship from domestic pharmaceutical companies is higher than that from overseas companies. Conclusions China has made significant advancements in diabetes drug research and development over the past decade. However, problems such as serious drug homogeneity, and insufficient innovation have become increasingly prominent. The government, clinical trial institutions, and pharmaceutical companies must collaborate to promote the high-quality development of drug clinical trials.
Collapse
Affiliation(s)
- Zengqing Ma
- Department of Pharmacy, Nanjing Gaochun People’s Hospital, Nanjing, Jiangsu, China
- Clinical Trial Center, Nanjing Gaochun People’s Hospital, Nanjing, Jiangsu, China
| | - Xin Zhao
- Department of Pharmacy, Nanjing Gaochun People’s Hospital, Nanjing, Jiangsu, China
| | - Yu Lin
- Department of Respiratory Medicine, Nanjing Gaochun People’s Hospital, Nanjing, Jiangsu, China
| | - Hong Zhang
- Department of Pharmacy, Nanjing Gaochun People’s Hospital, Nanjing, Jiangsu, China
- Clinical Trial Center, Nanjing Gaochun People’s Hospital, Nanjing, Jiangsu, China
| | - Lianping Wu
- Department of Pharmacy, Nanjing Gaochun People’s Hospital, Nanjing, Jiangsu, China
| | - Yang Tao
- Department of Pharmacy, Nanjing Gaochun People’s Hospital, Nanjing, Jiangsu, China
- Clinical Trial Center, Nanjing Gaochun People’s Hospital, Nanjing, Jiangsu, China
| | - Hongjun Shi
- Department of Pharmacy, Nanjing Gaochun People’s Hospital, Nanjing, Jiangsu, China
- Clinical Trial Center, Nanjing Gaochun People’s Hospital, Nanjing, Jiangsu, China
| | - Susu Li
- Department of Pharmacy, Nanjing Gaochun People’s Hospital, Nanjing, Jiangsu, China
| |
Collapse
|
27
|
Jiang Z, Kadeerhan G, Zhang J, Guo W, Guo H, Wang D. Advances in prostate-specific membrane antigen-targeted theranostics: from radionuclides to near-infrared fluorescence technology. Front Immunol 2025; 15:1533532. [PMID: 39867892 PMCID: PMC11757288 DOI: 10.3389/fimmu.2024.1533532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 12/13/2024] [Indexed: 01/28/2025] Open
Abstract
Prostate-Specific Membrane Antigen (PSMA) is a highly expressed and structurally unique target specific to prostate cancer (PCa). Diagnostic and therapeutic approaches in nuclear medicine, coupling PSMA ligands with radionuclides, have shown significant clinical success. PSMA-PET/CT effectively identifies tumors and metastatic lymph nodes for imaging purposes, while 177Lu-PSMA-617 (Pluvicto) has received FDA approval for treating metastatic castration-resistant PCa (mCRPC). Despite their success, radionuclide-based diagnostic and therapeutic methods face limitations such as high costs and significant side effects. Recently, near-infrared (NIR) fluorescence imaging and phototherapy have advanced significantly in biomedical applications. It's benefits, such as deep tissue penetration, real-time precision, and minimal side effects, have driven broader clinical adoption, especially in fluorescence-guided surgery (FGS). This review suggests combining NIR dyes with PSMA ligands to enable targeted, high-resolution imaging with superior signal-to-background ratios, facilitating precise FGS. NIR techniques can also aid pathological diagnosis in ex vivo specimens. Furthermore, combining photosensitizers with PSMA ligands allows localized photothermal (PTT) or photodynamic therapy (PDT) under NIR irradiation, producing heat or reactive oxygen species (ROS) to treat PCa. This review aims to extend the clinical success of radionuclide-based PSMA targeting by exploring advances in NIR-based FGS and phototherapy, presenting a promising new diagnostic and therapeutic approach.
Collapse
Affiliation(s)
- Zhongji Jiang
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Gaohaer Kadeerhan
- Central Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Jin Zhang
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wenmin Guo
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Hong Guo
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Dongwen Wang
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
28
|
Pandey SK, Sabharwal U, Tripathi S, Mishra A, Yadav N, Dwivedi-Agnihotri H. Androgen Signaling in Prostate Cancer: When a Friend Turns Foe. Endocr Metab Immune Disord Drug Targets 2025; 25:37-56. [PMID: 38831575 DOI: 10.2174/0118715303313528240523101940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/17/2024] [Accepted: 05/02/2024] [Indexed: 06/05/2024]
Abstract
Androgen (AR) signaling is the main signaling for the development of the prostate and its normal functioning. AR is highly specific for testosterone and dihydrotestosterone, significantly contributing to prostate development, physiology, and cancer. All these receptors have emerged as crucial therapeutic targets for PCa. In the year 1966, the Noble prize was awarded to Huggins and Hodge for their groundbreaking discovery of AR. As it is a pioneer transcription factor, it belongs to the steroid hormone receptor family and consists of domains, including DNA binding domain (DBD), hormone response elements (HRE), C-terminal ligand binding domain (LBD), and N-terminal regulatory domains. Structural variations in AR, such as AR gene amplification, LBD mutations, alternative splicing of exons, hypermethylation of AR, and co- regulators, are major contributors to PCa. It's signaling is crucial for the development and functioning of the prostate gland, with the AR being the key player. The specificity of AR for testosterone and dihydrotestosterone is important in prostate physiology. However, when it is dysregulated, AR contributes significantly to PCa. However, the structural variations in AR, such as gene amplification, mutations, alternative splicing, and epigenetic modifications, drive the PCa progression. Therefore, understanding AR function and dysregulation is essential for developing effective therapeutic strategies. Thus, the aim of this review was to examine how AR was initially pivotal for prostate development and how it turned out to show both positive and detrimental implications for the prostate.
Collapse
Affiliation(s)
- Swaroop Kumar Pandey
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura, 281406, India
| | - Usha Sabharwal
- P. G. Department of Biosciences, Centre of Advanced Studies, Satellite Campus, Sardar Patel Maidan, 388120, Gujarat, India
| | - Swati Tripathi
- Section of Electron Microscopy, Supportive Centre for Brain Research, National Institute for Physiological Sciences (NIPS) Okazaki, 444-8787, Japan
| | - Anuja Mishra
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura, 281406, India
| | - Neha Yadav
- Department of Biophysics, University of Delhi, South Campus, New Delhi, 110021, India
| | | |
Collapse
|
29
|
Wang X, Wang M, Lin Q, He L, Zhang B, Chen X, Chen G, Du H, Lang C, Peng X, Dai Y. Osteoblast-Derived ECM1 Promotes Anti-Androgen Resistance in Bone Metastatic Prostate Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407662. [PMID: 39563492 PMCID: PMC11727142 DOI: 10.1002/advs.202407662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 10/14/2024] [Indexed: 11/21/2024]
Abstract
Acquired resistance to hormonal therapy, particularly enzalutamide (ENZ), remains a significant obstacle in the treatment of advanced bone metastatic prostate cancer. Here, it is demonstrated that under ENZ treatment, osteoblasts in the bone microenvironment secrete increased levels of extracellular matrix protein 1 (ECM1), which affects surrounding prostate cancer cells, promoting tumor cell proliferation and anti-androgen resistance. Mechanistically, ECM1 interacts with the enolase 1 (ENO1) receptor on the prostate cancer cell membrane, leading to its phosphorylation at the Y189 site. This event further recruits adapter proteins including growth factor receptor-bound protein 2 (GRB2) and son of sevenless homolog 1 (SOS1), which activates the downstream mitogen-activated protein kinase (MAPK) signaling pathway to induce anti-androgen resistance. Furthermore, inhibiting ECM1 or utilizing the ENO1-targeting inhibitor phosphonoacetohydroxamate (PhAH) significantly restores tumor cell sensitivity to ENZ. Taken together, a potential mechanism is identified through which osteoblast-derived ECM1 drives resistance in bone metastatic prostate cancer under ENZ treatment. Additionally, the findings indicate that ECM1 and ENO1 may serve as potential targets for developing therapies for bone metastatic castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Xinwen Wang
- Department of Orthopedic Surgerythe First Affiliated HospitalSun Yat‐Sen UniversityGuangzhou510080China
- Guangdong Provincial Key Laboratory of Orthopedics and TraumatologyGuangzhou510080China
| | - Min Wang
- Guangdong Provincial Key Laboratory of Orthopedics and TraumatologyGuangzhou510080China
- Department of PathologyGuangzhou First People's HospitalGuangzhou510080China
| | - Qijun Lin
- Department of Orthopedic Surgerythe First Affiliated HospitalSun Yat‐Sen UniversityGuangzhou510080China
- Guangdong Provincial Key Laboratory of Orthopedics and TraumatologyGuangzhou510080China
| | - Lixin He
- Department of Experimental ResearchState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Baolin Zhang
- Department of Orthopedic Surgerythe First Affiliated HospitalSun Yat‐Sen UniversityGuangzhou510080China
- Guangdong Provincial Key Laboratory of Orthopedics and TraumatologyGuangzhou510080China
| | - Xin Chen
- Department of Orthopedic Surgerythe First Affiliated HospitalSun Yat‐Sen UniversityGuangzhou510080China
- Guangdong Provincial Key Laboratory of Orthopedics and TraumatologyGuangzhou510080China
| | - Guanhong Chen
- Department of Orthopedic Surgerythe First Affiliated HospitalSun Yat‐Sen UniversityGuangzhou510080China
- Guangdong Provincial Key Laboratory of Orthopedics and TraumatologyGuangzhou510080China
| | - Hong Du
- Department of PathologyGuangzhou First People's HospitalGuangzhou510080China
| | - Chuandong Lang
- Department of OrthopedicsThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230001China
| | - Xinsheng Peng
- Department of Orthopedic Surgerythe First Affiliated HospitalSun Yat‐Sen UniversityGuangzhou510080China
- Guangdong Provincial Key Laboratory of Orthopedics and TraumatologyGuangzhou510080China
| | - Yuhu Dai
- Department of Orthopedic Surgerythe First Affiliated HospitalSun Yat‐Sen UniversityGuangzhou510080China
- Guangdong Provincial Key Laboratory of Orthopedics and TraumatologyGuangzhou510080China
| |
Collapse
|
30
|
Zhu D, Pan Y, Yang Y, Wang S. Regulation of the Cilia as a Potential Treatment for Senescence and Tumors: A Review. J Cell Physiol 2025; 240:e31499. [PMID: 39660388 DOI: 10.1002/jcp.31499] [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: 07/16/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 12/12/2024]
Abstract
Millions of people worldwide die from malignant tumors every year, and the current clinical treatment is still based on radiotherapy and chemotherapy. Immunotherapy-adjuvant chemotherapy is widely applied, yet resistance to various factors persists in the management of advanced malignancies. Recently researchers have gradually discovered that the integrity of primary cilia is closely related to many diseases. The phenotypic changes in primary cilia are found in some cases of progeria, tumorigenesis, and drug resistance. Primary cilia seem to mediate signaling during these diseases. Hedgehog inhibitors have emerged in recent years to treat tumors by controlling signaling proteins on primary cilia. There is evidence for the use of anti-tumor drugs to treat senescence-related disease. Considering the close relationship between aging and obesity, as well as the obesity is the phenotype of many ciliopathies. Therefore, we speculate that some anti-tumor or anti-aging drugs can treat ciliopathies. Additionally, there is evidence suggesting that anti-aging drugs for tumor treatment, in which the process may be mediated by cilia. This review elucidates for the first time that cilia may be involved in the regulation of senescence, metabolic, tumorigenesis, and tumor resistance and hypothesizes that cilia can be regulated to treat these diseases in the future.
Collapse
Affiliation(s)
- Danping Zhu
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuqin Pan
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yong Yang
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Shukui Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Collaborative Innovation Center on Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
31
|
Bakht MK, Beltran H. Biological determinants of PSMA expression, regulation and heterogeneity in prostate cancer. Nat Rev Urol 2025; 22:26-45. [PMID: 38977769 PMCID: PMC11841200 DOI: 10.1038/s41585-024-00900-z] [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] [Accepted: 05/21/2024] [Indexed: 07/10/2024]
Abstract
Prostate-specific membrane antigen (PSMA) is an important cell-surface imaging biomarker and therapeutic target in prostate cancer. The PSMA-targeted theranostic 177Lu-PSMA-617 was approved in 2022 for men with PSMA-PET-positive metastatic castration-resistant prostate cancer. However, not all patients respond to PSMA-radioligand therapy, in part owing to the heterogeneity of PSMA expression in the tumour. The PSMA regulatory network is composed of a PSMA transcription complex, an upstream enhancer that loops to the FOLH1 (PSMA) gene promoter, intergenic enhancers and differentially methylated regions. Our understanding of the PSMA regulatory network and the mechanisms underlying PSMA suppression is evolving. Clinically, molecular imaging provides a unique window into PSMA dynamics that occur on therapy and with disease progression, although challenges arise owing to the limited resolution of PET. PSMA regulation and heterogeneity - including intertumoural and inter-patient heterogeneity, temporal changes, lineage dynamics and the tumour microenvironment - affect PSMA theranostics. PSMA response and resistance to radioligand therapy are mediated by a number of potential mechanisms, and complementary biomarkers beyond PSMA are under development. Understanding the biological determinants of cell surface target regulation and heterogeneity can inform precision medicine approaches to PSMA theranostics as well as other emerging therapies.
Collapse
Affiliation(s)
- Martin K Bakht
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
32
|
Zhong C, Wang J, Peng H, Lu J, Long Z, Lin Z, Chen G, Cai C, Cheng S, Chen Z, Zhang L, Zhong W, Mo R, Mao X. GG-NER's role in androgen receptor signaling inhibitor response for advanced prostate cancer. Cell Commun Signal 2024; 22:600. [PMID: 39696559 DOI: 10.1186/s12964-024-01977-0] [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: 09/05/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Advanced prostate cancer (PCa) often initially responds to androgen receptor signaling inhibitors (ARSI) but frequently develops resistance, driven by tumor heterogeneity and therapeutic pressure. Addressing the clinical challenge of identifying non-responsive patients and discovering new therapeutic targets is urgently needed. METHODS We utilized single-sample gene set enrichment analysis (ssGSEA) to elucidate the influence of the GG-NER pathway on ARSI response in PCa. We then constructed and validated a prognostic model based on this pathway using LASSO regression, Kaplan-Meier analysis, Cox regression, and ROC analysis. Additionally, we mapped tumor mutations to delineate the mutational landscapes across different risk groups and explored functional pathways through GO, KEGG, and GSEA analyses. The impact of the GG-NER pathway on enzalutamide sensitivity and DNA repair in PCa was further validated through CCK-8 assays, colony formation assays, in vivo experiments, and immunofluorescence. RESULTS ssGSEA indicated a trend of GG-NER pathway upregulation in patients with poor ARSI response. The GG-NER characteristic gene score (NECGS) identified a high-risk group with diminished ARSI response, serving as an independent prognostic indicator with strong predictive power. This high-risk group exhibited elevated TP53 mutation frequencies and significant enrichment in key pathways such as ribosome and mitochondrial functions, as well as MYC and E2F signaling. Experimental validation confirmed that targeting the GG-NER pathway or its key gene, ACTL6A, significantly reduces enzalutamide resistance in resistant cell lines and increases γH2AX expression. CONCLUSION NECGS effectively predicts ARSI response in PCa, and our comprehensive analysis underscores the critical role of the GG-NER pathway in enzalutamide resistance, positioning ACTL6A as a potential therapeutic target for PCa.
Collapse
Affiliation(s)
- Chuanfan Zhong
- Department of Urology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, Guangdong, China
- Department of Urology, The Tenth Affiliated Hospital of Southern Medical University (Dongguan people's hospital), 523059, Dongguan, Guangdong, China
| | - Jiaxing Wang
- Department of Urology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, Guangdong, China
| | - Hangyang Peng
- Department of Urology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, Guangdong, China
| | - Jianming Lu
- Department of Andrology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China.
| | - Zining Long
- Department of Urology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, Guangdong, China
| | - Zhuoyuan Lin
- Department of Urology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Guo Chen
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Chao Cai
- Department of Urology, Minimally Invasive Surgery Center, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shilong Cheng
- Department of Urology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, Guangdong, China
| | - Zhongjie Chen
- Department of Urology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, Guangdong, China
| | - Le Zhang
- Institute for Integrative Genome Biology, University of California, Riverside, California, United States
| | - Weibo Zhong
- Department of Urology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, Guangdong, China.
| | - Rujun Mo
- Department of Urology, The Tenth Affiliated Hospital of Southern Medical University (Dongguan people's hospital), 523059, Dongguan, Guangdong, China.
| | - Xiangming Mao
- Department of Urology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, Guangdong, China.
| |
Collapse
|
33
|
Celada SI, Li G, Celada LJ, Kanagasabai T, Lu W, Brown LK, Mark ZA, Izban MG, Ballard BR, Zhou X, Adunyah SE, Matusik RJ, Wang X, Chen Z. Castration-resistant prostate cancer is resensitized to androgen deprivation by autophagy-dependent apoptosis induced by blocking SKP2. Sci Signal 2024; 17:eadk4122. [PMID: 39689183 PMCID: PMC11784317 DOI: 10.1126/scisignal.adk4122] [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: 08/20/2023] [Revised: 08/04/2024] [Accepted: 11/18/2024] [Indexed: 12/19/2024]
Abstract
Resistance to androgen receptor (AR)-targeted therapies for prostate cancer (PCa) is characteristic of an aggressive subtype called castration-resistant prostate cancer (CRPC) and is often associated with tumor relapse. Both relapse and poor prognosis in patients with CRPC are associated with increased abundance of the E3 ubiquitin ligase SKP2. Therefore, we investigated the therapeutic potential of combined inhibition of AR and SKP2 for CRPC. We found that combined targeting of AR and SKP2 with small-molecule inhibitors decreased proliferation in two CRPC cell lines in culture and in xenografts in humanized mice. Furthermore, combined therapy in mice markedly decreased the growth of Pten/Trp53 double-knockout tumors, a particularly invasive model of CRPC, whereas disruption of either AR or SKP2 alone only modestly suppressed their growth. Mechanistically, the inhibition of SKP2 in CRPC cells induced autophagy-dependent apoptosis and promoted luminal-associated phenotypes, which promoted responsiveness to AR-targeted therapy. These effects were further enhanced by coinhibition of AR and were not induced by the AR inhibitor alone. Our findings indicate that targeting both AR and SKP2 signaling pathways is necessary to treat CRPC.
Collapse
Affiliation(s)
- Sherly I. Celada
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN 37208, USA
- Department of Biological Sciences, Tennessee State University, Nashville, TN 37209, USA
| | - Guoliang Li
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN 37208, USA
| | - Lindsay J. Celada
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Thanigaivelan Kanagasabai
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN 37208, USA
| | - Wenfu Lu
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN 37208, USA
| | - LaKendria K. Brown
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN 37208, USA
| | - Zaniya A. Mark
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN 37208, USA
| | - Michael G. Izban
- Department of Pathology, Anatomy and Cell Biology, Meharry Medical College, Nashville, TN 37208, USA
| | - Billy R. Ballard
- Department of Pathology, Anatomy and Cell Biology, Meharry Medical College, Nashville, TN 37208, USA
| | - Xinchun Zhou
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Samuel E. Adunyah
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN 37208, USA
| | - Robert J. Matusik
- Department of Urology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Xiaofei Wang
- Department of Biological Sciences, Tennessee State University, Nashville, TN 37209, USA
| | - Zhenbang Chen
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN 37208, USA
| |
Collapse
|
34
|
Madikonda AK, Ajayakumar A, Nadendla S, Banothu J, Muripiti V. Esterase-responsive nanoparticles (ERN): A targeted approach for drug/gene delivery exploits. Bioorg Med Chem 2024; 116:118001. [PMID: 39556942 DOI: 10.1016/j.bmc.2024.118001] [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: 09/02/2024] [Revised: 10/29/2024] [Accepted: 11/08/2024] [Indexed: 11/20/2024]
Abstract
Nanoparticles are being developed to enhance drug delivery to cancer tumors, leveraging advantages such as the enhanced permeability and retention (EPR) effect. However, traditional nanoparticles often face challenges with low specificity for cancer cells, leading to inefficient delivery and unwanted side effects. Esterase-responsive nanoparticles offer a maximum targeted approach to tumor cells because they release their therapeutic payload at the tumor site under the influence of esterase activity. This review explores the role of esterase-responsive nanoparticles in drug and gene delivery, examines esterase prodrug therapy, and discusses prostate-specific membrane antigen (PSMA) targets esterase-responsive nanoparticles in prostate cancer treatment. Additionally, we reviewed the current research progress and future potential of esterase-responsive nanoparticles in enhancing drug and gene delivery.
Collapse
Affiliation(s)
- Ashok Kumar Madikonda
- Department of Biochemistry & Molecular Biology, Central University of Kerala, Tejaswini Hills, Periye, Kasaragod 671320, Kerala, India
| | - Amritha Ajayakumar
- Department of Biochemistry & Molecular Biology, Central University of Kerala, Tejaswini Hills, Periye, Kasaragod 671320, Kerala, India
| | - Sudeena Nadendla
- Department of Chemistry, Central University of Kerala, Tejaswini Hills, Periye, Kasaragod 671320, Kerala, India
| | - Janardhan Banothu
- Department of Chemistry, National Institute of Technology Calicut, Kozhikode 673601, Kerala, India
| | - Venkanna Muripiti
- Department of Education, Central University of Kerala, Tejaswini Hills, Periye, Kasaragod 671320, Kerala, India.
| |
Collapse
|
35
|
Wu L, Wang J, Chai L, Chen J, Jin X. Roles of deubiquitinases in urologic cancers (Review). Oncol Lett 2024; 28:609. [PMID: 39525605 PMCID: PMC11544529 DOI: 10.3892/ol.2024.14743] [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: 07/23/2024] [Accepted: 09/23/2024] [Indexed: 11/16/2024] Open
Abstract
Human health is endangered by the occurrence and progression of urological cancers, including renal cell carcinoma, prostate cancer and bladder cancer, which are usually associated with the activation of oncogenic factors and inhibition of cancer suppressors. The primary mechanism for protein breakdown in cells is the ubiquitin-proteasome system, whilst deubiquitinases contribute to the reversal of this process. However, both are important for protein homeostasis. Deubiquitination may also be involved in the control of the cell cycle, proliferation and apoptosis, and dysregulated deubiquitination is associated with the malignant transformation, invasion and metastasis of urologic malignancies. Therefore, a comprehensive summary of the mechanisms underlying deubiquitination in urological cancers may provide novel strategies and insights for diagnosis and treatment. The present review aimed to methodically clarify the role of deubiquitinating enzymes in urinary system cancers as well as their prospective application prospects for clinical treatment.
Collapse
Affiliation(s)
- Liangpei Wu
- Department of Chemoradiotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Jiahui Wang
- Department of Chemoradiotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Lin Chai
- Department of Chemoradiotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Jun Chen
- Department of Chemoradiotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| |
Collapse
|
36
|
Ou Y, Xia C, Ye C, Liu M, Jiang H, Zhu Y, Yang D. Comprehensive scRNA-seq analysis to identify new markers of M2 macrophages for predicting the prognosis of prostate cancer. Ann Med 2024; 56:2398195. [PMID: 39221762 PMCID: PMC11370685 DOI: 10.1080/07853890.2024.2398195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 05/06/2024] [Accepted: 05/20/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Prostate cancer (PCa) has become the highest incidence of malignant tumor among men in the world. Tumor microenvironment (TME) is necessary for tumor growth. M2 macrophages play an important role in many solid tumors. This research aimed at the role of M2 macrophages' prognosis value in PCa. METHODS Single-cell RNA-seq (scRNA-seq) data and mRNA expression data were obtained from the Gene Expression Omnibus database (GEO) and The Cancer Genome Atlas (TCGA). Quality control, normalization, reduction, clustering, and cell annotation of scRNA-seq data were preformed using the Seruat package. The sub-populations of the tumor-associated macrophages (TAMs) were analysis and the marker genes of M2 macrophage were selected. Differentially expressed genes (DEGs) in PCa were identified using limma and the immune infiltration was detected using CIBERSORTx. Then, a weighted correlation network analysis (WGCNA) was constructed to identify the M2 macrophage-related modules and genes. Integration of the marker genes of M2 macrophage from scRNA-seq data analysis and hub genes from WGCNA to select the prognostic gene signature based on Univariate and LASSO regression analysis. The risk score was calculated, and the DEGs, biological function, immune characteristics related to risk score were explored. And a predictive nomogram was constructed. CCK8, Transwell, and wound healing were used to verify cell phenotype changes after co-cultured. RESULTS A total of 2431 marker genes of M2 macrophage and 650 hub M2 macrophage-related genes were selected based on scRNA-seq data and WGCNA. Then, 113 M2 macrophage-related genes were obtained by overlapping the scRNA-seq data and WGCNA results. Nine M2 macrophage-related genes (SMOC2, PLPP1, HES1, STMN1, GPR160, ABCG1, MAZ, MYC, and EPCAM) were screened as prognostic gene signatures. M2 risk score was calculated, the DEGs, Immune score, stromal score, ESTIMATE score, tumor purity, and immune cell infiltration, immune checkpoint expression, and responses of immunotherapy and chemotherapy were identified. And a predictive nomogram was constructed. CCK8, Transwell invasion, and wound healing further verified that M2 macrophages promoted the proliferation, invasion, and migration of PCa (p < 0.05). CONCLUSIONS We uncovered that M2 macrophages and relevant genes played key roles in promoting the occurrence, development, and metastases of PCa and played as convincing predictors in PCa.
Collapse
Affiliation(s)
- Yitian Ou
- Urology Department, Kunming Medical University Second Affiliated Hospital, Kunming, Yunnan, China
| | - Chengxing Xia
- Urology Department, Kunming Medical University Second Affiliated Hospital, Kunming, Yunnan, China
| | - Chunwei Ye
- Urology Department, Kunming Medical University Second Affiliated Hospital, Kunming, Yunnan, China
| | - Mingming Liu
- Urology Department, Kunming Medical University Second Affiliated Hospital, Kunming, Yunnan, China
| | - Haiyang Jiang
- Urology Department, Kunming Medical University Second Affiliated Hospital, Kunming, Yunnan, China
| | - Yong Zhu
- Urology Department, Kunming Medical University Second Affiliated Hospital, Kunming, Yunnan, China
| | - Delin Yang
- Urology Department, Kunming Medical University Second Affiliated Hospital, Kunming, Yunnan, China
| |
Collapse
|
37
|
Qin X, Niu R, Tan Y, Huang Y, Ren W, Zhou W, Wu H, Zhang J, Xu M, Zhou X, Guan H, Zhu X, Chen Y, Cao K. Exosomal PSM-E inhibits macrophage M2 polarization to suppress prostate cancer metastasis through the RACK1 signaling axis. Biomark Res 2024; 12:138. [PMID: 39538297 PMCID: PMC11562865 DOI: 10.1186/s40364-024-00685-8] [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: 08/05/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND It is well-established that understanding the mechanism of prostate cancer (PCa)-associated metastasis is paramount for improving its prognosis. Metastasis is known to involve the communication between tumor-associated macrophages (TAMs) and tumor cells. Exosomes are crucial in mediating this intercellular communication within the tumor microenvironment. Nonetheless, the role of exosomal proteins in PCa metastasis is not yet fully understood. Here, we investigated the mechanisms of prostate cancer-derived exosomal PSM-E on regulating macrophage M2 polarization to suppress tumor invasion and metastasis. METHODS PSM-E levels in exosomes were detected by transmission electron microscopy and Western blotting analysis. The diagnostic value of urine-derived exosomal PSM-E in PCa were evaluated by LC-MS/MS, correlation analysis, and ROC curves analysis. The mechanisms underlying the inhibitory effect of exosomal PSM-E on the M2 polarization of macrophages was investigated by co-IP, IHC staining, and PCa tumorigenesis model, etc. RESULTS: We revealed that exosomal PSM-E is upregulated in exosomes derived from the serum and urine of PCa patients. Clinically, an elevated exosomal PSM-E expression in urine is significantly correlated with an advanced pathological tumor stage and a high Gleason score. Our research also revealed that exosomal PSM-E inhibits prostate cancer cell proliferation, invasion, and metastasis by suppressing macrophage polarization in vitro and in vivo. Furthermore, we provided compelling evidence that exosomal PSM-E inhibits M2 polarization of macrophages by recruiting RACK1 and suppressing the FAK and ERK signaling pathways, consequently suppressing PCa invasion and metastasis. Furthermore, we found that the protease-associated domain of PSM-E and the fourth tryptophan-aspartate repeat of RACK1 are crucial for the interaction between PSM-E and RACK1. CONCLUSIONS Notably, exosomes carrying PSM-E from PCa urine could potentially serve as a biomarker for PCa, and targeting exosomal PSM-E may represent a strategy for preventing tumor progression in this patient population.
Collapse
Affiliation(s)
- Xingliang Qin
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Ruoxi Niu
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Yongyao Tan
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
- Research Center for Clinical Laboratory Standard, Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan Road II, Guangzhou, 510080, China
| | - Yuxin Huang
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Weishu Ren
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
- Research Center for Clinical Laboratory Standard, Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan Road II, Guangzhou, 510080, China
| | - Weiwei Zhou
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
- Research Center for Clinical Laboratory Standard, Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan Road II, Guangzhou, 510080, China
| | - Huiquan Wu
- Guangzhou Jishiyuan Bio-technology Co., Ltd., Guangzhou, 510700, China
| | - Junlong Zhang
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Mingze Xu
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiang Zhou
- Department of Microsurgery, Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hongyu Guan
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Xun Zhu
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China.
- Research Center for Clinical Laboratory Standard, Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan Road II, Guangzhou, 510080, China.
| | - Yu Chen
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
| | - Kaiyuan Cao
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China.
- Research Center for Clinical Laboratory Standard, Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan Road II, Guangzhou, 510080, China.
- Guangzhou Jishiyuan Bio-technology Co., Ltd., Guangzhou, 510700, China.
| |
Collapse
|
38
|
Antar RM, Fawaz C, Gonzalez D, Xu VE, Drouaud AP, Krastein J, Pio F, Murdock A, Youssef K, Sobol S, Whalen MJ. The Evolving Molecular Landscape and Actionable Alterations in Urologic Cancers. Curr Oncol 2024; 31:6909-6937. [PMID: 39590142 PMCID: PMC11593205 DOI: 10.3390/curroncol31110511] [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: 10/20/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
The genetic landscape of urologic cancers has evolved with the identification of actionable mutations that impact diagnosis, prognosis, and therapeutic strategies. This narrative review consolidates existing literature on genetic mutations across key urologic cancers, including bladder, renal, prostate, upper tract urothelial, testicular, and penile. The review highlights mutations in DNA damage repair genes, such as BRCA1/2 and PTEN, as well as pathway alterations like FGFR and PD-L1 overexpression. These mutations influence tumor behavior and therapeutic outcomes, emphasizing the need for precision oncology approaches. Molecular profiling, through tools like next-generation sequencing, has revolutionized patient care by enabling targeted treatment strategies, especially in cancers with distinct molecular subtypes such as luminal or basal bladder cancer and clear cell renal carcinoma. Emerging therapies, including FGFR inhibitors and immune checkpoint blockade, offer new treatment avenues, although resistance mechanisms remain a challenge. We also emphasize the importance of biomarker identification for personalized management, especially in metastatic settings where treatment intensification is often required. Future research is needed to further elucidate our understanding of the genetics affecting urologic cancers, which will help develop novel, individualized therapies to enhance oncologic outcomes.
Collapse
Affiliation(s)
- Ryan Michael Antar
- Department of Urology, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA (M.J.W.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Hu J, Zhang J, Han B, Qu Y, Zhang Q, Yu Z, Zhang L, Han J, Liu H, Gao L, Feng T, Dou B, Chen W, Sun F. PLXNA1 confers enzalutamide resistance in prostate cancer via AKT signaling pathway. Neoplasia 2024; 57:101047. [PMID: 39226661 PMCID: PMC11419896 DOI: 10.1016/j.neo.2024.101047] [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: 07/13/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/05/2024]
Abstract
Although targeting the androgen signaling pathway by androgen receptor (AR) inhibitors, including enzalutamide, has shown therapeutic effectiveness, inevitable emergence of acquired resistance remains a critical challenge in the treatment of advanced prostate cancer (PCa). Recognizing targetable genomic aberrations that trigger endocrine treatment failure holds great promise for advancing therapeutic interventions. Here, we characterized PLXNA1, amplified in a subset of PCa patients, as a contributor to enzalutamide resistance (ENZR). Elevated PLXNA1 expression facilitated PCa proliferation under enzalutamide treatment due to AKT signaling activation. Mechanistically, PLXNA1 recruited NRP1 forming a PLXNA1-NRP1 complex, which in turn potentiated the phosphorylation of the AKT. Either inhibiting PLXNA1-NRP1 complex with an NRP1 inhibitor, EG01377, or targeting PLXNA1-mediated ENZR with AKT inhibitors, abolished the pro-resistance phenotype of PLXNA1. Taken together, combination of AKT inhibitor and AR inhibitors presents a promising therapeutic strategy for PCa, especially in advanced PCa patients exhibiting PLXNA1 overexpression.
Collapse
Affiliation(s)
- Jing Hu
- Department of Pathology, Qilu Hospital, Shandong University, Jinan 250012, China; Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Jing Zhang
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Bo Han
- Department of Pathology, Peking University People's Hospital, Beijing, China
| | - Ying Qu
- Department of Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Qian Zhang
- Department of Pathology, Binzhou Medical University Hospital, Binzhou 256603, China
| | - Zeyuan Yu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China
| | - Lin Zhang
- Yinzhou District Center for Disease Control and Prevention, Ningbo, China
| | - Jingying Han
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China
| | - Hui Liu
- Department of Pathology, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Lin Gao
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China
| | - Tingting Feng
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China
| | - Baokai Dou
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Weiwen Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Jinan 250012, China
| | - Feifei Sun
- Department of Pathology, Qilu Hospital, Shandong University, Jinan 250012, China.
| |
Collapse
|
40
|
Li Z, Li Z, Luo Y, Chen W, Fang Y, Xiong Y, Zhang Q, Yuan D, Yan B, Zhu J. Application and new findings of scRNA-seq and ST-seq in prostate cancer. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:23. [PMID: 39470950 PMCID: PMC11522250 DOI: 10.1186/s13619-024-00206-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 10/12/2024] [Indexed: 11/01/2024]
Abstract
Prostate cancer is a malignant tumor of the male urological system with the highest incidence rate in the world, which seriously threatens the life and health of middle-aged and elderly men. The progression of prostate cancer involves the interaction between tumor cells and tumor microenvironment. Understanding the mechanisms of prostate cancer pathogenesis and disease progression is important to guide diagnosis and therapy. The emergence of single-cell RNA sequencing (scRNA-seq) and spatial transcriptome sequencing (ST-seq) technologies has brought breakthroughs in the study of prostate cancer. It makes up for the defects of traditional techniques such as fluorescence-activated cell sorting that are difficult to elucidate cell-specific gene expression. This review summarized the heterogeneity and functional changes of prostate cancer and tumor microenvironment revealed by scRNA-seq and ST-seq, aims to provide a reference for the optimal diagnosis and treatment of prostate cancer.
Collapse
Affiliation(s)
- Zhuang Li
- Department of Urology, Affiliated Hospital of Guizhou Medical University, Guiyang city, 550004, Guizhou Province, China
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang city, 550002, Guizhou Province, China
| | - Zhengnan Li
- Graduate School of Zunyi Medical University, Zunyi City, 563099, Guizhou Province, China
| | - Yuanyuan Luo
- Medical College of Guizhou University, Guiyang city, 550025, Guizhou Province, China
| | - Weiming Chen
- Medical College of Guizhou University, Guiyang city, 550025, Guizhou Province, China
| | - Yinyi Fang
- Medical College of Guizhou University, Guiyang city, 550025, Guizhou Province, China
| | - Yuliang Xiong
- Department of Urology, Affiliated Hospital of Guizhou Medical University, Guiyang city, 550004, Guizhou Province, China
| | - Qinyi Zhang
- Graduate School of Zunyi Medical University, Zunyi City, 563099, Guizhou Province, China
| | - Dongbo Yuan
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang city, 550002, Guizhou Province, China
| | - Bo Yan
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang city, 550002, Guizhou Province, China
| | - Jianguo Zhu
- Department of Urology, Affiliated Hospital of Guizhou Medical University, Guiyang city, 550004, Guizhou Province, China.
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang city, 550002, Guizhou Province, China.
- Graduate School of Zunyi Medical University, Zunyi City, 563099, Guizhou Province, China.
- Medical College of Guizhou University, Guiyang city, 550025, Guizhou Province, China.
| |
Collapse
|
41
|
Qiu Y, Man C, Zhu L, Zhang S, Wang X, Gong D, Fan Y. R-loops' m6A modification and its roles in cancers. Mol Cancer 2024; 23:232. [PMID: 39425197 PMCID: PMC11487993 DOI: 10.1186/s12943-024-02148-y] [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: 07/02/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024] Open
Abstract
R-loops are three-stranded nucleic acid structures composed of an RNA-DNA hybrid and a displaced DNA strand. They are widespread and play crucial roles in regulating gene expression, DNA replication, and DNA and histone modifications. However, their regulatory mechanisms remain unclear. As R-loop detection technology advances, changes in R-loop levels have been observed in cancer models, often associated with transcription-replication conflicts and genomic instability. N6-methyladenosine (m6A) is an RNA epigenetic modification that regulates gene expression by affecting RNA localization, splicing, translation, and degradation. Upon reviewing the literature, we found that R-loops with m6A modifications are implicated in tumor development and progression. This article summarizes the molecular mechanisms and detection methods of R-loops and m6A modifications in gene regulation, and reviews recent research on m6A-modified R-loops in oncology. Our goal is to provide new insights into the origins of genomic instability in cancer and potential strategies for targeted therapy.
Collapse
Affiliation(s)
- Yue Qiu
- Cancer Institute, Affiliated People's Hospital of Jiangsu University, No 8, Dianli Road, Zhenjiang, Jiangsu Province, 212002, People's Republic of China
| | - Changfeng Man
- Cancer Institute, Affiliated People's Hospital of Jiangsu University, No 8, Dianli Road, Zhenjiang, Jiangsu Province, 212002, People's Republic of China
| | - Luyu Zhu
- Department of Gastroenterology, The Suqian Clinical College of Xuzhou Medical University, No 120, Suzhi Road, Suqian, Jiangsu Province, 223812, People's Republic of China
| | - Shiqi Zhang
- Department of Gastroenterology, The Suqian Clinical College of Xuzhou Medical University, No 120, Suzhi Road, Suqian, Jiangsu Province, 223812, People's Republic of China
| | - Xiaoyan Wang
- Department of Gastroenterology, The Suqian Clinical College of Xuzhou Medical University, No 120, Suzhi Road, Suqian, Jiangsu Province, 223812, People's Republic of China.
| | - Dandan Gong
- Cancer Institute, Affiliated People's Hospital of Jiangsu University, No 8, Dianli Road, Zhenjiang, Jiangsu Province, 212002, People's Republic of China.
| | - Yu Fan
- Cancer Institute, Affiliated People's Hospital of Jiangsu University, No 8, Dianli Road, Zhenjiang, Jiangsu Province, 212002, People's Republic of China.
| |
Collapse
|
42
|
Tabibian M, Moghaddam FS, Motevaseli E, Ghafouri-Fard S. Targeting mRNA-coding genes in prostate cancer using CRISPR/Cas9 technology with a special focus on androgen receptor signaling. Cell Commun Signal 2024; 22:504. [PMID: 39420406 PMCID: PMC11484332 DOI: 10.1186/s12964-024-01833-1] [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: 08/13/2024] [Accepted: 09/17/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Prostate cancer is among prevalent cancers in men. Numerous strategies have been proposed to intervene with the important prostate cancer-related signaling pathways. Among the most promising strategies is CRISPR/Cas9 strategy. This strategy has been used to modify expression of a number of genes in prostate cancer cells. AIMS This review summarizes the most recent progresses in the application of CRISPR/Cas9 strategy in modification of prostate cancer-related phenotypes with an especial focus on pathways related to androgen receptor signaling. CONCLUSION CRISPR/Cas9 technology has successfully targeted several genes in the prostate cancer cells. Moreover, the efficiency of this technique in reducing tumor burden has been tested in animal models of prostate cancer. Most of targeted genes have been related with the androgen receptor signaling. Targeted modulation of these genes have affected growth of castration-resistant prostate cancer. PI3K/AKT/mTOR signaling and immune response-related genes have been other targets that have been successfully modulated by CRISPR/Cas9 technology in prostate cancer. Based on the rapid translation of this technology into the clinical application, it is anticipated that novel treatments based on this technique change the outcome of this malignancy in future.
Collapse
Affiliation(s)
- Mobina Tabibian
- Department of Cellular and Molecular Biology, Faculty of Life Sciences and Biotechnologies, Shahid Beheshti University, Tehran, Iran
| | | | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
43
|
Zou C, Zhu J, Xiong J, Tian Y, Peng Y, Cheung E, Zhang D. Comprehensive Characterization of the Integrin Family Across 32 Cancer Types. GENOMICS, PROTEOMICS & BIOINFORMATICS 2024; 22:qzae035. [PMID: 39436262 PMCID: PMC11849494 DOI: 10.1093/gpbjnl/qzae035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 02/19/2024] [Accepted: 05/06/2024] [Indexed: 10/23/2024]
Abstract
Integrin genes are widely involved in tumorigenesis. Yet, a comprehensive characterization of integrin family members and their interactome at the pan-cancer level is lacking. Here, we systematically analyzed integrin family in approximately 10,000 tumors across 32 cancer types. Globally, integrins represent a frequently altered and misexpressed pathway, with alteration and dysregulation overall being protumorigenic. Expression dysregulation, better than mutational landscape, of integrin family successfully identifies a subgroup of aggressive tumors with a high level of proliferation and stemness. The results reveal that several molecular mechanisms collectively regulate integrin expression in a context-dependent manner. For potential clinical usage, we constructed a weighted scoring system, integrinScore, to measure integrin signaling patterns in individual tumors. Remarkably, integrinScore was consistently correlated with predefined molecular subtypes in multiple cancers, with integrinScore-high tumors being more aggressive. Importantly, integrinScore was cancer-dependent and closely associated with proliferation, stemness, tumor microenvironment, metastasis, and immune signatures. IntegrinScore also predicted patients' response to immunotherapy. By mining drug databases, we unraveled an array of compounds that may modulate integrin signaling. Finally, we built a user-friendly database, Pan-cancer Integrin Explorer (PIExplorer; http://computationalbiology.cn/PIExplorer), to facilitate researchers to explore integrin-related knowledge. Collectively, we provide a comprehensive characterization of integrins across cancers and offer gene-specific and cancer-specific rationales for developing integrin-targeted therapy.
Collapse
Affiliation(s)
- Cheng Zou
- Hunan Key Laboratory of Animal Models and Molecular Medicine, School of Biomedical Sciences, Hunan University, Changsha 410082, China
| | - Jinwei Zhu
- Hunan Key Laboratory of Animal Models and Molecular Medicine, School of Biomedical Sciences, Hunan University, Changsha 410082, China
| | - Jiangling Xiong
- Hunan Key Laboratory of Animal Models and Molecular Medicine, School of Biomedical Sciences, Hunan University, Changsha 410082, China
| | - Yu Tian
- Hunan Key Laboratory of Animal Models and Molecular Medicine, School of Biomedical Sciences, Hunan University, Changsha 410082, China
| | - Yousong Peng
- College of Biology, Hunan University, Changsha 410082, China
| | - Edwin Cheung
- Faculty of Health Sciences, University of Macau, Macau Special Administrative Region 999078, China
| | - Dingxiao Zhang
- Hunan Key Laboratory of Animal Models and Molecular Medicine, School of Biomedical Sciences, Hunan University, Changsha 410082, China
| |
Collapse
|
44
|
Yu J, Zhou K, Wang J, Mao L. Preliminary Efficacy, Tolerability, and Safety Analysis of Darolutamide for Metastatic Castration-Resistant Prostate Cancer: A Single-Center, Open-Label Study. Urol Int 2024; 109:167-174. [PMID: 39406197 DOI: 10.1159/000541929] [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: 05/07/2024] [Accepted: 09/23/2024] [Indexed: 11/15/2024]
Abstract
INTRODUCTION Darolutamide is a structurally unique second-generation androgen receptor antagonist that has been approved for indications in patients with non-metastatic castration-resistant prostate cancer (nmCRPC) and metastatic hormone-sensitive prostate cancer (mHSPC). The aim was to assess the efficacy and safety of Darolutamide for mCRPC. METHODS In this single-center, open-label study, patients with previously untreated mCRPC were enrolled and received androgen deprivation therapy (goserelin acetate 3.6 mg every 28 days) and docetaxel (75 mg per square meter of body surface area every 21 days) with Denosumab (120 mg every 28 days) for bone metastases, Darolutamide (300 mg orally twice daily) in the experimental group, and the control group received the corresponding of placebo. Serum PSA changes were detected and recorded, and imaging changes and adverse events (AEs) were evaluated. The primary endpoints were safety, tolerability, and antitumor efficacy, and the second endpoint was radiographic progression-free survival (rPFS). RESULTS Thirty-seven patients with mCRPC were enrolled. The median time to PSA50 in the Darolutamide group was 1.5 months (95% CI: 0.2619-0.9545), significantly lower than that in the placebo group (3.0 months [95% CI: 1.048-3.818], p = 0.0259). The median time to PSA90 in the experimental group was 4 months (95% CI: 0.3094-1.437), 2 months shorter than that in the placebo group (6.0 months [95% CI: 0.6961-3.232]). With the median follow-up of 6 months, the median decrease in serum PSA was -81.8% (range -60.4 to -99.9%) in the Darolutamide group and -69.4% (range -50.3 to -89.6%) in the placebo group. Tumor-related pain and AEs were not increased, and the median rPFS was not reached. CONCLUSIONS The combination of Darolutamide and docetaxel was well tolerated with more clinically beneficial than docetaxel alone in previously untreated mCRPC. Darolutamide rapidly reduced PSA levels and prolonged rPFS and did not increase the incidence of AEs.
Collapse
Affiliation(s)
- JunJie Yu
- Department of Urology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China,
| | - KaiChen Zhou
- Department of Urology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - JunQi Wang
- Department of Urology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - LiJun Mao
- Department of Urology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
45
|
Okpako IO, Ng'ong'a FA, Kyama CM, Njeru SN. Network pharmacology, molecular docking, and in vitro study on Aspilia pluriseta against prostate cancer. BMC Complement Med Ther 2024; 24:338. [PMID: 39304868 DOI: 10.1186/s12906-024-04642-8] [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: 11/06/2023] [Accepted: 09/11/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Current prostate cancer treatments are associated with life-threatening side effects, prompting the search for effective and safer alternatives. Aspilia pluriseta Schweinf. ex Engl. has previously shown anticancer activity in lung and liver cancer cell lines. This study investigated its potential for prostate cancer. METHODS A crude extract of A. pluriseta root was prepared using dichloromethane/methanol (1:1 v/v) and partitioned into hexane, ethyl acetate, and water fractions. The MTT assay was used to assess the antiproliferative activity of the fractions. The active fractions were tested at 6.25-200 µg/ml on human prostate cancer DU-145 cells and non-cancerous Vero E6 cells. Qualitative phytochemical and gas chromatography-mass spectrometry (GC-MS) analyses were conducted to identify chemical compounds. Network pharmacology was employed to predict molecular targets and modes of action of the identified chemical compounds, with subsequent validation through molecular docking and real-time PCR. RESULTS Active extracts included crude dichloromethane/methanol, hexane, and ethyl acetate fractions, inhibiting DU-145 cell proliferation with IC50 values of 16.94, 20.06, and 24.14 µg/ml, respectively. Selectivity indices were determined to be 6.04 (crude), 3.62 (hexane), and 6.68 (ethyl acetate). Identified phytochemicals comprised phenols, terpenoids, flavonoids, tannins, sterols, and saponins. GC-MS analysis revealed seventy-nine (79) compounds, with seven (7) meeting ideal drug candidate parameters; their hub gene targets included MAPK3, MAPK1, IL6, TP53, ESR1, PTGS2, MMP9, MDM2, AR, and MAP2K1, implicating regulation of PI3K/Akt, MAPK, and p53 signaling pathways as potential modes of action. Core compounds such as 1-heneicosanol, lanosterol, andrographolide, and retinoic acid exhibited strong binding activities, particularly lanosterol with MAPK21 (-9.7 kcal/mol), ESR1 (-8.9 kcal/mol), and MAPK3 (-8.8 kcal/mol). Treatment with A. pluriseta downregulated AR expression and upregulated p53, while also downregulating CDK1 and BCL-2 and upregulating caspase-3. CONCLUSIONS A. pluriseta extracts inhibited DU-145 cell growth without causing cellular toxicity, suggesting great potential for development as an anti-prostate cancer agent. However, further in vitro and in vivo experiments are recommended.
Collapse
Affiliation(s)
- Innocent Oluwaseun Okpako
- Department of Molecular Biology and Biotechnology, Pan African University Institute for Basic Sciences, Technology and Innovation, Nairobi, Kenya.
| | - Florence Atieno Ng'ong'a
- Department of Biochemistry, School of Biomedical Sciences, College of Health Sciences, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Cleophas Mutinda Kyama
- Department of Medical Laboratory Sciences, School of Biomedical Sciences, College of Health Sciences, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Sospeter Ngoci Njeru
- Centre for Traditional Medicine and Drug Research and Centre for Community Driven Research, Kenya Medical Research Institute, Nairobi, Kenya.
| |
Collapse
|
46
|
Kwon WA, Song YS, Lee MK. Strategic Advances in Combination Therapy for Metastatic Castration-Sensitive Prostate Cancer: Current Insights and Future Perspectives. Cancers (Basel) 2024; 16:3187. [PMID: 39335158 PMCID: PMC11430187 DOI: 10.3390/cancers16183187] [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: 08/23/2024] [Revised: 09/14/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024] Open
Abstract
The contemporary treatment for metastatic castration-sensitive prostate cancer (mCSPC) has evolved significantly, building on successes in managing metastatic castration-resistant prostate cancer (mCRPC). Although androgen deprivation therapy (ADT) alone has long been the cornerstone of mCSPC treatment, combination therapies have emerged as the new standard of care based on recent advances, offering improved survival outcomes. Landmark phase 3 trials demonstrated that adding chemotherapy (docetaxel) and androgen receptor pathway inhibitors to ADT significantly enhances overall survival, particularly for patients with high-volume, high-risk, or de novo metastatic disease. Despite these advancements, a concerning gap between evidence-based guidelines and real-world practice remains, with many patients not receiving recommended combination therapies. The challenge in optimizing therapy sequences, considering both disease control and treatment burdens, and identifying clinical and biological subgroups that could benefit from personalized treatment strategies persists. The advent of triplet therapy has shown promise in extending survival, but the uro-oncology community must narrow the gap between evidence and practice to deliver the most effective care. Current research is focused on refining treatment approaches and utilizing biomarkers to guide therapy selection, aiming to offer more personalized and adaptive strategies for mCSPC management. Thus, aligning clinical practices with the evolving evidence is urgently needed to improve outcomes for patients facing this incurable disease.
Collapse
Affiliation(s)
- Whi-An Kwon
- Department of Urology, Hanyang University College of Medicine, Myongji Hospital, Goyang 10475, Republic of Korea
| | - Yong Sang Song
- Department of Obstetrics and Gynecology, Hanyang University College of Medicine, Myongji Hospital, Goyang 10475, Republic of Korea
| | - Min-Kyung Lee
- Department of Internal Medicine, Hanyang University College of Medicine, Myongji Hospital, Goyang 10475, Republic of Korea
| |
Collapse
|
47
|
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
|
48
|
Kunath F, Heidegger I, Heck M, Mayr R, Becker C, Rausch S. [Molecular tumor boards in uro-oncology-prostate cancer]. UROLOGIE (HEIDELBERG, GERMANY) 2024; 63:899-907. [PMID: 39107624 DOI: 10.1007/s00120-024-02399-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/27/2024] [Indexed: 08/24/2024]
Abstract
The rapid development of molecular medicine has opened up new perspectives for the diagnosis and treatment of urological tumors. Urology faces the challenge of effectively treating advanced cancer, especially in view of the genetic diversity of urological tumors. The molecular tumor board offers an innovative approach to identify targeted therapy options based on the individual genetic signatures of tumor cells or tumor microenvironment-based treatment options. In this article, the concept of the molecular tumor board in urology is presented using the example of prostate cancer. We discuss the principles, applications, and future prospects of this promising approach.
Collapse
Affiliation(s)
- Frank Kunath
- Klinik für Urologie und Kinderurologie, Klinikum Bayreuth GmbH, Bayreuth, Deutschland
- Fachgruppe Molekulare Urologie, Arbeitsgruppe urologische Forschung (AuF), Deutsche Gesellschaft für Urologie e. V., Düsseldorf, Deutschland
| | - Isabel Heidegger
- Abteilung für Urologie, Medizinische Universität Innsbruck, Innsbruck, Österreich
- Fachgruppe Molekulare Urologie, Arbeitsgruppe urologische Forschung (AuF), Deutsche Gesellschaft für Urologie e. V., Düsseldorf, Deutschland
| | - Matthias Heck
- Klinik und Poliklinik für Urologie, Klinikum rechts der Isar, Technische Universität München, München, Deutschland
- Fachgruppe Molekulare Urologie, Arbeitsgruppe urologische Forschung (AuF), Deutsche Gesellschaft für Urologie e. V., Düsseldorf, Deutschland
| | - Roman Mayr
- Klinik für Urologie, Caritas-Krankenhaus St. Josef, Universität Regensburg, Regensburg, Deutschland
- Fachgruppe Molekulare Urologie, Arbeitsgruppe urologische Forschung (AuF), Deutsche Gesellschaft für Urologie e. V., Düsseldorf, Deutschland
| | - Christoph Becker
- Ressort Forschungsförderung, Deutsche Gesellschaft für Urologie e. V., Düsseldorf, Deutschland.
- Fachgruppe Molekulare Urologie, Arbeitsgruppe urologische Forschung (AuF), Deutsche Gesellschaft für Urologie e. V., Düsseldorf, Deutschland.
- Forschungskoordination, Geschäftsstelle Düsseldorf, Deutsche Gesellschaft für Urologie e. V., Uerdinger Str. 64, 40474, Düsseldorf, Deutschland.
| | - Steffen Rausch
- Klinik für Urologie, Universitätsklinik Tübingen, Tübingen, Deutschland
- Fachgruppe Molekulare Urologie, Arbeitsgruppe urologische Forschung (AuF), Deutsche Gesellschaft für Urologie e. V., Düsseldorf, Deutschland
| |
Collapse
|
49
|
Cao Y, Jia M, Duan C, Yang Z, Cheng B, Wang R. The m 6A regulators in prostate cancer: molecular basis and clinical perspective. Front Pharmacol 2024; 15:1448872. [PMID: 39268470 PMCID: PMC11391310 DOI: 10.3389/fphar.2024.1448872] [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: 06/14/2024] [Accepted: 08/14/2024] [Indexed: 09/15/2024] Open
Abstract
Prostate cancer (PCa) is the second leading cause of cancer-related death among men in western countries. Evidence has indicated the significant role of the androgen receptor (AR) as the main driving factor in controlling the development of PCa, making androgen receptor inhibition (ARI) therapy a pivotal management approach. In addition, AR independent signaling pathways also contribute to PCa progression. One such signaling pathway that has garnered our attention is N6-Methyladenosine (m6A) signaling, which refers to a chemical modification on RNA with crucial roles in RNA metabolism and disease progression, including PCa. It is important to comprehensively summarize the role of each individual m6A regulator in PCa development and understand its interaction with AR signaling. This review aims to provide a thorough summary of the involvement of m6A regulators in PCa development, shedding light on their upstream and downstream signaling pathways. This summary sets the stage for a comprehensive review that would benefit the scientific community and clinical practice by enhancing our understanding of the biology of m6A regulators in the context of PCa.
Collapse
Affiliation(s)
- Yu Cao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Man Jia
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Chunyan Duan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Zhihui Yang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Bo Cheng
- Department of Urology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Ronghao Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| |
Collapse
|
50
|
Thakur N, Singh P, Bagri A, Srivastava S, Dwivedi V, Singh A, Jaiswal SK, Dholpuria S. Therapy resistance in prostate cancer: mechanism, signaling and reversal strategies. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:1110-1134. [PMID: 39351434 PMCID: PMC11438573 DOI: 10.37349/etat.2024.00266] [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: 05/17/2024] [Accepted: 08/09/2024] [Indexed: 10/04/2024] Open
Abstract
Prostate cancer (PC) depicts a major health challenge all over the globe due to its complexities in the treatment and diverse clinical trajectories. Even in the advances in the modern treatment strategies, the spectrum of resistance to the therapies continues to be a significant challenge. This review comprehensively examines the underlying mechanisms of the therapy resistance occurred in PC, focusing on both the tumor microenvironment and the signaling pathways implicated in the resistance. Tumor microenvironment comprises of stromal and epithelial cells, which influences tumor growth, response to therapy and progression. Mechanisms such as microenvironmental epithelial-mesenchymal transition (EMT), anoikis suppression and stimulation of angiogenesis results in therapy resistance. Moreover, dysregulation of signaling pathways including androgen receptor (AR), mammalian target of rapamycin/phosphoinositide 3 kinase/AKT (mTOR/PI3K/AKT), DNA damage repair and Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathways drive therapy resistance by promoting tumor survival and proliferation. Understanding these molecular pathways is important for developing targeted therapeutic interventions which overcomes resistance. In conclusion, a complete grasp of mechanisms and pathways underlying medication resistance in PC is important for the development of individualized treatment plans and enhancements of clinical outcomes. By studying and understanding the complex mechanisms of signaling pathways and microenvironmental factors contributing to therapy resistance, this study focuses and aims to guide the development of innovative therapeutic approaches to effectively overcome the PC progression and improve the survival rate of patients.
Collapse
Affiliation(s)
- Neha Thakur
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand 248002, India
| | - Pallavi Singh
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand 248002, India
| | - Aditi Bagri
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand 248002, India
| | - Saumya Srivastava
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand 248002, India
| | - Vinay Dwivedi
- Amity Institute of Biotechnology, Amity University, Gwalior, Madhya Pradesh 474005, India
| | - Asha Singh
- Amity Institute of Biotechnology, Amity University, Gwalior, Madhya Pradesh 474005, India
| | - Sunil Kumar Jaiswal
- School of Biological and Life Sciences, Galgotias University, Greater Noida, Uttar Pradesh 203201, India
| | - Sunny Dholpuria
- Department of Life Sciences, J. C. Bose University of Science and Technology, YMCA Faridabad, Faridabad, Haryana 121006, India
| |
Collapse
|