1
|
Cai S, Deng Y, Zou Z, Tian W, Tang Z, Li J, Tan Z, Wu Z, Han Z, Wen B, Feng Y, Liu R, Zhu X, Wu Y, Xiao H, He H, Ye J, Zhong W. Metformin inhibits the progression of castration-resistant prostate cancer by regulating PDE6D induced purine metabolic alternation and cGMP / PKG pathway activation. Cancer Lett 2025; 622:217694. [PMID: 40216151 DOI: 10.1016/j.canlet.2025.217694] [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: 02/26/2025] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 05/01/2025]
Abstract
The castration-resistant prostate cancer (CRPC) remains an incurable disease. Metformin has demonstrated a potential therapeutic effect on CRPC. However, the poor clinical performance of metformin against cancer may be due to its clinical dose being much lower than the anticancer concentration used in pre-clinical experiments. The challenge is to determine a way to enhance sensitivity to metformin at an appropriate concentration on CRPC. In this study, a mouse model of low-dose metformin treatment for CRPC cells were established. Metabolomic-seq and transcriptomic-seq was used to investigate changes in CRPC xenografts. We discovered that low-dose metformin inhibits the progression of CRPC by regulating PDE6D, which induces alterations in purine metabolism and activates the cGMP/PKG pathway. Furthermore, we found that cells with high expression of PDE6D were more resistant to metformin. When combined with the PDE6D inhibitor TMX-4100, the inhibitory effect on tumors was enhanced, and TMX-4100 demonstrated favorable biosafety in animal models. In conclusion, we found that low-dose metformin inhibits the progression of CRPC by regulating PDE6D-induced alterations in purine metabolism and activating the cGMP/PKG pathway. Moreover, patients with high PDE6D expression may exhibit greater resistance to metformin. Combining metformin with TMX-4100 could further improve the inhibitory effects on tumors.
Collapse
Affiliation(s)
- Shanghua Cai
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China; Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, 510005, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, 510230, Guangzhou, Guangdong, China
| | - Yulin Deng
- Department of Urology, The First Dongguan Affiliated Hospital, Guangdong Medical University, 523710, Dongguan, Guangdong, China
| | - Zhihao Zou
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China; Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, 510005, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, 510230, Guangzhou, Guangdong, China
| | - Weicheng Tian
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, 510230, Guangzhou, Guangdong, China
| | - Zhenfeng Tang
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China; Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, 510005, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, 510230, Guangzhou, Guangdong, China
| | - Jinchuang Li
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China; Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China
| | - Zeheng Tan
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China; Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China
| | - Zhenjie Wu
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China; Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China
| | - Zhaodong Han
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China; Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China
| | - Biyan Wen
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, 510230, Guangzhou, Guangdong, China
| | - Yuanfa Feng
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, 510230, Guangzhou, Guangdong, China
| | - Ren Liu
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China
| | - Xuejin Zhu
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China
| | - Yongding Wu
- Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China
| | - Haiyin Xiao
- Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China
| | - Huichan He
- Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, 510230, Guangzhou, Guangdong, China.
| | - Jianheng Ye
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, 999078, Macao Special Administrative Region of China; Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China.
| | - Weide Zhong
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China; Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, 510005, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, 510230, Guangzhou, Guangdong, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, 999078, Macao Special Administrative Region of China; Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China.
| |
Collapse
|
2
|
Abida W, Beltran H, Raychaudhuri R. State of the Art: Personalizing Treatment for Patients With Metastatic Castration-Resistant Prostate Cancer. Am Soc Clin Oncol Educ Book 2025; 45:e473636. [PMID: 40112242 DOI: 10.1200/edbk-25-473636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Until recently, the treatment of metastatic castration-resistant prostate cancer (mCRPC) relied exclusively on hormonal therapies and taxane chemotherapy. The advent of modern molecular profiling methods applied in the clinic, namely, next-generation sequencing and advanced positron emission tomography (PET) imaging, has allowed for the development of biomarker-driven therapeutics including anti-PD-L1 therapy for microsatellite instability-high or tumor mutation burden-high disease, poly(ADP-ribose) polymerase (PARP) inhibitors for patients with DNA damage repair mutations, and lutetium 177 vipivotide tetraxetan (177Lu-PSMA-617) for patients with prostate-specific membrane antigen (PSMA) PET-avid disease. While these targeted therapies have improved outcomes, there is an opportunity to refine biomarkers to optimize patient selection, understand resistance, and develop novel combination strategies. In addition, studies in the laboratory and in patient-derived samples have shown that a subset of mCRPC tumors lose expression of common prostate cancer markers such as prostate-specific antigen and PSMA because of lineage plasticity and the development of non-androgen receptor (AR)-driven disease. Non-AR-driven prostate cancer has been associated with aggressive behavior and poor prognosis, including in some cases histologic transformation to a poorly differentiated neuroendocrine prostate cancer (NEPC). The clinical management of NEPC typically follows the treatment paradigm for small cell lung cancer and increasingly relies on genomic and phenotypic characterization of disease, including loss of tumor suppressors and expression of cell surface markers such as DLL3. Therefore, both genomic subtyping and phenotypic subtyping are important to consider and can guide the clinical management of patients with advanced prostate cancer.
Collapse
Affiliation(s)
- Wassim Abida
- Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Ruben Raychaudhuri
- University of Washington and the Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
3
|
Navin AK, Rejani CT, Chandrasekaran B, Tyagi A. Urolithins: Emerging natural compound targeting castration-resistant prostate cancer (CRPC). Biomed Pharmacother 2025; 187:118058. [PMID: 40253830 DOI: 10.1016/j.biopha.2025.118058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/03/2025] [Accepted: 04/14/2025] [Indexed: 04/22/2025] Open
Abstract
Castration-resistant prostate cancer (CRPC) presents a significant challenge due to its resistance to conventional androgen deprivation therapies. Urolithins, bioactive metabolites derived from ellagitannins, have recently emerged as promising therapeutic agents for CRPC. Urolithins not only inhibit androgen receptor (AR) signaling, a crucial factor in the progression of CRPC, but also play a key role in regulating oxidative stress by their antioxidant properties, thereby inhibiting increased reactive oxygen species, a common feature of the aggressive nature of CRPC. Research has shown that urolithins induce apoptosis and diminish pro-survival signaling, leading to tumor inhibition. This review delves into the intricate mechanisms through which urolithins exert their therapeutic effects, focusing on both AR-dependent and AR-independent pathways. It also explores the exciting potential of combining urolithins with androgen ablation therapy, opening new avenues for CRPC treatment.
Collapse
Affiliation(s)
- Ajit Kumar Navin
- Department of Pharmacology, College of Pharmacy, Texas A&M University, College Station, TX 77845, USA
| | | | - Balaji Chandrasekaran
- Department of Pharmacology, College of Pharmacy, Texas A&M University, College Station, TX 77845, USA
| | - Ashish Tyagi
- Department of Pharmacology, College of Pharmacy, Texas A&M University, College Station, TX 77845, USA.
| |
Collapse
|
4
|
Adzavon YM, Culig Z, Sun Z. Interactions between androgen and IGF1 axes in prostate tumorigenesis. Nat Rev Urol 2025; 22:268-275. [PMID: 39375467 DOI: 10.1038/s41585-024-00942-3] [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: 09/04/2024] [Indexed: 10/09/2024]
Abstract
Androgen signalling through the androgen receptor (AR) is essential for prostate tumorigenesis. However, androgen signalling pathways also interact with other growth factor-mediated signalling pathways to regulate the prostatic cell cycle, differentiation, apoptosis and proliferation in the initiation and progression of prostate cancer. Insulin-like growth factor 1 (IGF1) is one of the most prominent growth factors in prostate tumorigenesis. Clinical and experimental evidence has demonstrated that IGF1 signalling supports both androgen-dependent and androgen-independent prostate tumorigenesis, suggesting that improved understanding of the interactions between the IGF1 and androgen axes might aid the development of new therapeutic strategies. Available data have shown a dynamic role of androgen-AR signalling in the activation of IGF1-signalling pathways by augmenting transcription of the IGF1 receptor in prostatic basal epithelial cells and by increasing IGF1 secretion through the suppression of IGF-binding protein 3 expression in prostatic stromal cells. In turn, IGF1 stimulates Wnt-β-catenin signalling in prostatic basal progenitors to promote prostatic oncogenic transformation and prostate cancer development. These findings highlight the cooperative, autocrine and paracrine interactions that underlie the oncogenic effects of androgens and IGF1 and open up new opportunities for therapeutic targeting.
Collapse
Affiliation(s)
- Yao Mawulikplimi Adzavon
- Department of Cell Biology, Montefiore Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Oncology, Montefiore Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Zoran Culig
- Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Zijie Sun
- Department of Cell Biology, Montefiore Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
5
|
Durán AM, Whitley K, Santiago K, Yoo C, Valdez G, Cheng KW, Ochoa P, de Semir D, Xiu J, Chokkalingam P, Das S, Schaefer ES, Rowe SP, Das BC, Casiano CA, Almaguel F. Inhibition of Mitochondrial-Associated Protein MAGMAS Resensitizes Chemoresistant Prostate Cancer Cells to Docetaxel. Cancers (Basel) 2025; 17:1535. [PMID: 40361461 PMCID: PMC12072152 DOI: 10.3390/cancers17091535] [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: 03/19/2025] [Revised: 04/15/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES Metastatic prostate cancer (PCa) is the leading cause of cancer-related deaths and a major contributor to cancer mortality in men. Most patients with metastatic PCa eventually develop metastatic castration-resistant prostate cancer (mCRPC), characterized by resistance to treatment with androgen-deprivation therapy, and often later the development of resistance to other types of agents. MAGMAS, a 13.8 kDa mitochondrial-associated protein, facilitates the import of nuclear-encoded proteins into the mitochondrial matrix. Overexpression of MAGMAS has been observed in several aggressive cancers, including breast, glioblastoma, and prostate cancer. When overexpressed, MAGMAS acts as a cytoprotective protein by scavenging reactive oxygen species (ROS), maintaining ROS levels that support cell proliferation while avoiding the induction of apoptosis. This study investigates the role of MAGMAS in therapy resistance in PCa cells. METHODS/RESULTS Quantitative immunoblotting revealed that MAGMAS is endogenously upregulated in docetaxel-resistant (DR) PCa cell lines compared to their docetaxel-sensitive parental counterparts. While MAGMAS depletion alone did not affect the survival of DR cells, it significantly sensitized them to docetaxel (DTX), as indicated by a marked reduction in clonogenic potential. Additionally, transient knockdown of MAGMAS in these resistant cells significantly decreased the levels of ABCB1 protein. Consistent with these findings, sub-therapeutic inhibition of MAGMAS using the novel BT#9 inhibitor, in combination with increasing concentrations of DTX, enhanced the sensitivity of DR cells to DTX, as demonstrated by proliferation and clonogenic assays. Lastly, RNA tumor expression predicts overall survival (OS). CONCLUSIONS These results implicate MAGMAS in PCa chemoresistance and suggest that targeting this protein could provide a novel therapeutic strategy for treating DR tumors.
Collapse
Affiliation(s)
- Alfonso M. Durán
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (K.S.); (C.Y.); (K.W.C.); (P.O.); (C.A.C.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
- Cancer Center, Loma Linda University Health, Loma Linda, CA 92354, USA
| | - Kristen Whitley
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (K.S.); (C.Y.); (K.W.C.); (P.O.); (C.A.C.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Krystal Santiago
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (K.S.); (C.Y.); (K.W.C.); (P.O.); (C.A.C.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Christian Yoo
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (K.S.); (C.Y.); (K.W.C.); (P.O.); (C.A.C.)
| | - Giancarlo Valdez
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (K.S.); (C.Y.); (K.W.C.); (P.O.); (C.A.C.)
| | - Kai Wen Cheng
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (K.S.); (C.Y.); (K.W.C.); (P.O.); (C.A.C.)
| | - Pedro Ochoa
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (K.S.); (C.Y.); (K.W.C.); (P.O.); (C.A.C.)
| | - David de Semir
- Caris Life Sciences, Phoenix, AZ 85040, USA; (D.d.S.); (J.X.)
| | - Joanne Xiu
- Caris Life Sciences, Phoenix, AZ 85040, USA; (D.d.S.); (J.X.)
| | - Parthiban Chokkalingam
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14215, USA; (P.C.); (S.D.); (B.C.D.)
| | - Sasmita Das
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14215, USA; (P.C.); (S.D.); (B.C.D.)
| | | | - Steven P. Rowe
- Department of Radiology, University of North Carolina, Chapel Hill, NC 27514, USA;
| | - Bhaskar C. Das
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14215, USA; (P.C.); (S.D.); (B.C.D.)
| | - Carlos A. Casiano
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (K.S.); (C.Y.); (K.W.C.); (P.O.); (C.A.C.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
- Cancer Center, Loma Linda University Health, Loma Linda, CA 92354, USA
- Department of Medicine, Rheumatology Division, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Frankis Almaguel
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (K.S.); (C.Y.); (K.W.C.); (P.O.); (C.A.C.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
- Cancer Center, Loma Linda University Health, Loma Linda, CA 92354, USA
| |
Collapse
|
6
|
Zhuang R, Xie R, Peng S, Zhou Q, Lin W, Ou Y, Chen B, Su T, Li Z, Huang H, Li K, Duan Y. An anti-androgen resistance-related gene signature acts as a prognostic marker and increases enzalutamide efficacy via PLK1 inhibition in prostate cancer. J Transl Med 2025; 23:480. [PMID: 40289088 PMCID: PMC12034143 DOI: 10.1186/s12967-025-06457-8] [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: 10/12/2024] [Accepted: 04/06/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Anti-androgen resistance remains a major clinical challenge in the treatment of prostate cancer (PCa), leading to disease progression and treatment failure. Despite extensive research on resistance mechanisms, a reliable prognostic model for predicting patient outcomes and guiding therapeutic strategies is still lacking. This study aimed to develop a novel gene signature related to anti-androgen resistance and evaluate its prognostic and therapeutic implications. METHODS Anti-androgen resistance-related differentially expressed genes (ARRDEGs) were identified through transcriptomic analysis of enzalutamide- and dual enzalutamide abiraterone-resistant PCa cell lines from the GEO database. Functional enrichment analysis was performed to determine the biological roles of these genes. A prognostic gene signature was developed using univariate Cox regression, LASSO, and multivariate Cox regression models. The model was validated in independent PCa cohorts from The Cancer Genome Atlas (TCGA). Additionally, we assessed the correlation between the signature, immune infiltration, immune checkpoint expression, and drug sensitivity. The efficacy of PLK1 inhibition combined with enzalutamide was further explored using in vitro and in vivo experiments. RESULTS We identified 304 ARRDEGs, from which three key genes (LMNB1, SSPO, and PLK1) were selected to construct a prognostic signature. This gene signature effectively stratified PCa patients into high- and low-risk groups, with the high-risk group exhibiting shorter recurrence-free survival and distinct immune characteristics. High-risk patients demonstrated elevated immune checkpoint expression (B7H3, CTLA-4, B7-1, and TIGIT), increased M2 macrophage infiltration, and enhanced sensitivity to chemotherapy and targeted therapy. Mechanistically, PLK1 inhibition potentiated the antitumor effect of enzalutamide by downregulating SLC7A11 and inducing ferroptosis, providing a potential therapeutic strategy to overcome anti-androgen resistance. CONCLUSION We established a novel ARRDEGs-based prognostic signature that predicts PCa progression and response to chemotherapy and targeted therapy. The integration of this signature with immune profiling and drug sensitivity analysis provides a valuable tool for precision oncology in PCa. Our findings highlight the potential of PLK1 inhibition as a therapeutic strategy to enhance enzalutamide efficacy and overcome resistance.
Collapse
Affiliation(s)
- Ruilin Zhuang
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Ruihui Xie
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Shirong Peng
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Qianghua Zhou
- Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Weilong Lin
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yuan Ou
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Bingliang Chen
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Tong Su
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Zean Li
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Hai Huang
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Department of Urology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China.
| | - Kaiwen Li
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| | - Yu Duan
- Department of Medical Ultrasonics, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China.
| |
Collapse
|
7
|
Perimbeti S, Jamroze A, Gopalakrishnan D, Jain R, Jiang C, Holleran JL, Parise RA, Bies R, Quinn D, Attwood K, Liu X, Kirk JS, Beumer JH, Tang DG, Chatta G. Phase Ib Study of Enzalutamide with Venetoclax in Patients with Metastatic Castration-Resistant Prostate Cancer. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.22.25326208. [PMID: 40313289 PMCID: PMC12045430 DOI: 10.1101/2025.04.22.25326208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Purpose Castration and enzalutamide induce BCL-2 to drive therapy resistance in prostate cancer (PCa). We conducted a phase Ib trial to test that metastatic castration-resistant PCa (mCRPC) can be effectively targeted by combining enzalutamide with the BCL-2 inhibitor venetoclax. Experimental Design This phase Ib single-arm trial of enzalutamide (160 mg/d) with venetoclax in patients with progressive mCRPC assessed dose-limiting toxicity (DLT), maximum tolerated dose (MTD) and recommended phase 2 dose (RP2D). Three dose levels (DL) of venetoclax (DL1 400 mg/d; DL2 600 mg/d; and DL3 800 mg) were evaluated using a 3+3 design. We also analyzed enzalutamide and venetoclax pharmacokinetics and conducted pharmacodynamic studies in peripheral blood mononuclear cells (PBMCs) to determine the impact of venetoclax on BCL-2 expression. Results A total of 10 patients were enrolled across 3 DL and no DLT was observed. Mean duration on treatment was 29 weeks (range: 8-140 weeks). Treatment-related adverse events (TRAEs) were mostly grade 1-2, and Grade 3 TRAEs included hypertension (20%), fatigue (10%), and thrombocytopenia (10%). 1/10 (10%) attained PSA50 response and 4/10 (40%) had stable disease. Estimated median overall survival (OS) was 19 months (95% CI 5-28 months) and median time to next systemic therapy (TNST) was 5 months (95% CI 1-35 months). Pharmacokinetic results revealed sub-therapeutic plasma levels of venetoclax. Pharmacodynamic studies demonstrated that venetoclax enhanced BCL-2β generation and promoted BCL-2 degradation. Conclusions Enzalutamide with venetoclax has an acceptable toxicity profile in patients with mCRPC. Despite sub-therapeutic venetoclax levels, the treatment elicited pharmacodynamic and clinical response in a subset of patients.
Collapse
Affiliation(s)
- Stuthi Perimbeti
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Anmbreen Jamroze
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | | | - Rohit Jain
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Changchuan Jiang
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Julianne L Holleran
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Robert A. Parise
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Robert Bies
- Department of Pharmaceutical Sciences, University at Buffalo School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
- Institute for Computational and Data Sciences, University at Buffalo, State University of New York at Buffalo, Buffalo, NY, USA
| | | | - Kristopher Attwood
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Xiaozhuo Liu
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Jason S. Kirk
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Jan H. Beumer
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Division of Hematology/Oncology, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, Pittsburgh, PA, USA
| | - Dean G. Tang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Experimental Therapeutics Graduate Program, University at Buffalo and Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Gurkamal Chatta
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
8
|
Guo J, Li N, Liu Q, Hao Z, Zhu G, Wang X, Wang H, Pan Q, Xu B, Han Y, Zhang G, Lian Y, Zhang W, Gu Y, Lin N, Zeng X, Jin Z, Lan W, Jiang J, Gao D, Dong L, Yuan H, Liang C, Qin J. KMT2C deficiency drives transdifferentiation of double-negative prostate cancer and confer resistance to AR-targeted therapy. Cancer Cell 2025:S1535-6108(25)00139-4. [PMID: 40280125 DOI: 10.1016/j.ccell.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 01/25/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025]
Abstract
Double-negative prostate cancer (DNPC), characterized by an androgen receptor (AR)- and neuroendocrine-null phenotype, frequently emerges following androgen deprivation therapy (ADT). However, our understanding of the origins and regulatory mechanisms of DNPC remains limited. Here, we discover that tumors with KMT2C mutation or loss are highly susceptible to transitioning into DNPC following ADT. We clarify that DNPC primarily stems from luminal cell transdifferentiation rather than basal cell transformation. Antiandrogen treatment induces KMT2C binding at enhancers of a subset of AR-regulated genes, preserving the adenocarcinoma lineage. KMT2C maintains ASPP2 expression via enhancer-promoter communication post-AR inhibition, while its inactivation reduces ASPP2, triggering ΔNp63-dependent transdifferentiation. This DNPC transition maintains fatty acid (FA) synthesis through ΔNp63-mediated SREBP1c transactivation, fueling DNPC growth via HRAS palmitoylation and MAPK signaling activation. These findings highlight KMT2C as an epigenetic checkpoint against DNPC development and suggest the therapeutic potential of targeting fatty acid synthesis.
Collapse
Affiliation(s)
- Jiacheng Guo
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Ni Li
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Jinfeng Laboratory, Chongqing 401329, China.
| | - Qiuli Liu
- Department of Urology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Zongyao Hao
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230022, China
| | - Guanghui Zhu
- West China School of Public Health, West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610000, China
| | - Xuege Wang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Hanling Wang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Qiang Pan
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Jinfeng Laboratory, Chongqing 401329, China
| | - Beitao Xu
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Ying Han
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Jinfeng Laboratory, Chongqing 401329, China
| | - Guoying Zhang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Yannan Lian
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Wei Zhang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Yongqiang Gu
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Naiheng Lin
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Xin Zeng
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Zige Jin
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Weihua Lan
- Department of Urology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Jun Jiang
- Department of Urology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Dong Gao
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Liang Dong
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| | - Huairui Yuan
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Chaozhao Liang
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230022, China.
| | - Jun Qin
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Jinfeng Laboratory, Chongqing 401329, China.
| |
Collapse
|
9
|
Shen J, Lu L, Chen Z, Guo W, Wang S, Liu Z, Gong X, Qi Y, Jin R, Zhang C. Multi-omics analysis constructs a novel neuroendocrine prostate cancer classifier and classification system. Sci Rep 2025; 15:13901. [PMID: 40263498 PMCID: PMC12015331 DOI: 10.1038/s41598-025-96683-3] [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: 12/30/2024] [Accepted: 03/31/2025] [Indexed: 04/24/2025] Open
Abstract
Neuroendocrine prostate cancer (NEPC), a subtype of prostate cancer (PCa) with poor prognosis and high heterogeneity, currently lacks accurate markers. This study aims to identify a robust NEPC classifier and provide new perspectives for resolving intra- tumoral heterogeneity. Multi-omics analysis included 19 bulk transcriptomics, 14 single-cell transcriptomics, 1 spatial transcriptomics, 16 published NE signatures and 10 cellular experiments combined with multiple machine learning algorithms to construct a novel NEPC classifier and classification. A comprehensive single-cell atlas of prostate cancer was created from 70 samples, comprising 196,309 cells, among which 9% were identified as NE cells. Within this framework and in combination with bulk transcriptomics, a total of 100 high-quality NE-specific feature genes were identified and differentiated into NEPup sig and NEPdown sig. The random forest (RF) algorithm proved to be the most effective classifier for NEPC, leading to the establishment of the NEP100 model, which demonstrated robust validation across various datasets. In clinical settings, the use of the NEP100 model can greatly improve the diagnostic and prognostic prediction of NEPC. Hierarchical clustering based on NEP100 revealed four distinct NEPC subtypes, designated VR_O, Prol_N, Prol_P, and EMT_Y, each of which presented unique biological characteristics. This allows us to select different targeted therapeutic strategies for different subtypes of phenotypic pathways. Notably, NEP100 expression correlated positively with neuroendocrine differentiation and disease progression, while the VR-NE phenotype dominated by VR_O cells indicated a propensity for treatment resistance. Furthermore, AMIGO2, a component of the NEP100 signature, was associated with chemotherapy resistance and a poor prognosis, indicating that it is a pivotal target for future therapeutic strategies. This study used multi-omics analysis combined with machine learning to construct a novel NEPC classifier and classification system. NEP100 provides a clinically actionable framework for NEPC diagnosis and subtyping.
Collapse
Affiliation(s)
- Junxiao Shen
- Department of Urology, The Fourth Affiliated Hospital of the School of Medicine, International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Luyuan Lu
- Department of General Surgery, The Fourth Affiliated Hospital of the School of Medicine, International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Zujie Chen
- Department of Urology, The Fourth Affiliated Hospital of the School of Medicine, International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Wei Guo
- Department of Urology, The Fourth Affiliated Hospital of the School of Medicine, International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Shuwen Wang
- Department of Urology, The Fourth Affiliated Hospital of the School of Medicine, International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Ziqiao Liu
- Department of Urology, The Fourth Affiliated Hospital of the School of Medicine, International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Xuke Gong
- Department of Urology, The Fourth Affiliated Hospital of the School of Medicine, International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Yiming Qi
- Department of Urology, The Fourth Affiliated Hospital of the School of Medicine, International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Ruyi Jin
- Department of Dermatology, NHC Key Laboratory of Immunodermatology, The First Hospital of China Medical University, China Medical University, Shenyang, 110001, People's Republic of China
| | - Cheng Zhang
- Department of Urology, The Fourth Affiliated Hospital of the School of Medicine, International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
| |
Collapse
|
10
|
Tang DG. Serotonin sets up neutrophil extracellular traps to promote neuroendocrine prostate cancer metastasis in the liver. J Clin Invest 2025; 135:e191687. [PMID: 40231471 PMCID: PMC11996856 DOI: 10.1172/jci191687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025] Open
Abstract
Castration-resistant prostate cancer frequently metastasizes to the liver, and prostate cancer liver metastases often present a neuroendocrine phenotype (i.e., neuroendocrine prostate cancer [NEPC]), but the underlying molecular underpinnings remain unclear. In this issue of the JCI, Liu et al. demonstrate that the neurotransmitter serotonin (also known as 5-hydroxytryptamine), produced by NEPC cells, gained access to and activated neutrophils by modifying histone 3 (H3) to form neutrophil extracellular traps, which in turn promoted NEPC macrometastases in the liver. The study suggests that blocking serotonin transport to neutrophils and inhibiting the enzymes that catalyze serotonin-mediated H3 modifications may represent alternative approaches to treating prostate cancer liver metastases.
Collapse
Affiliation(s)
- Dean G. Tang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- Experimental Therapeutics Graduate Program, University at Buffalo and Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| |
Collapse
|
11
|
Nørgaard M, Rusan M, Kondrup K, Sørensen EMG, Weiss S, Bjerre MT, Fredsøe J, Vang S, Jensen JB, De Laere B, Grönberg H, Borre M, Lindberg J, Sørensen KD. Deep targeted sequencing of circulating tumor DNA to inform treatment in patients with metastatic castration-resistant prostate cancer. J Exp Clin Cancer Res 2025; 44:120. [PMID: 40229848 PMCID: PMC11998381 DOI: 10.1186/s13046-025-03356-0] [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: 10/14/2024] [Accepted: 03/04/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Intrinsic and acquired resistance to second-generation anti-androgens pose a significant clinical challenge in the treatment of metastatic castration-resistant prostate cancer (mCRPC). Novel biomarkers to predict treatment response and inform alternative treatment options are urgently needed. METHODS Deep targeted sequencing, with a prostate cancer-specific gene panel, was performed on circulating tumor DNA (ctDNA) and germline DNA from blood of mCRPC patients recruited in Denmark (n = 53), prior to starting first-line treatment with enzalutamide or abiraterone acetate, and for a subset of patients also at progression (n = 18). Likely clonal hematopoietic variants were filtered out. Genomic findings were correlated to clinical outcomes (PSA progression-free survival (PFS), overall survival (OS)). Intrinsic resistance candidate biomarkers were considered by enrichment analysis of nonresponders vs. responders. Genomic alterations at progression were considered as possible drivers of acquired resistance. Clinical actionability was assessed based on OncoKB and ESCAT. RESULTS Somatic alterations in PTEN, cell cycle regulators (CCND1, CDKN1B, CDKN2A, and RB1) and chromatin modulators (CHD1, ARID1A) were associated with significantly shorter PFS and OS, also after adjusting for ctDNA% in multivariate Cox regression analysis. The associations with poorer outcomes for alterations in PTEN and chromatin modulators were validated in an external dataset. Patients with primary resistance to enzalutamide/abiraterone had enrichment for BRAF amplification and CHD1 loss, while responders had enrichment for TMPRSS2 fusions. AR resistance mutations emerged in 22% of patients at progression. These were mutually exclusive with other alterations that may confer resistance (i.e., activating CTNNB1 mutations, combined TP53/RB1 loss). Clinically actionable alterations, primarily in homologous recombination repair genes, were found in 54.7% and 49.0% of patients (OncoKB and ESCAT, respectively), with few additional alterations detected at progression. Level I alterations were identified in 41.5% of patients employing OncoKB, however only in 13.2% based on ESCAT. CONCLUSIONS Our study identifies known and novel prognostic and predictive biomarker candidates in patients with mCRPC undergoing first-line treatment with enzalutamide or abiraterone acetate. It further provides real-world evidence of the significant potential of genomic profiling of ctDNA to inform treatment in this setting. Clinical trials are warranted to advance the implementation of ctDNA-based biomarkers into clinical practice.
Collapse
Affiliation(s)
- Maibritt Nørgaard
- Department of Molecular Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus, 8200, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Maria Rusan
- Department of Molecular Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus, 8200, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus, Denmark
| | - Karoline Kondrup
- Department of Molecular Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus, 8200, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Ea Marie Givskov Sørensen
- Department of Molecular Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus, 8200, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Simone Weiss
- Department of Molecular Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus, 8200, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Marianne Trier Bjerre
- Department of Molecular Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus, 8200, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Urology, Aarhus University Hospital, Aarhus, Denmark
- Department of Urology, Gødstrup Hospital, Gødstrup, Denmark
| | - Jacob Fredsøe
- Department of Molecular Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus, 8200, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Søren Vang
- Department of Molecular Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus, 8200, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jørgen Bjerggaard Jensen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Urology, Gødstrup Hospital, Gødstrup, Denmark
| | - Bram De Laere
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Cancer Research Institute Gent (CRIG), Ghent University, Ghent, Belgium
| | - Henrik Grönberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Michael Borre
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Urology, Aarhus University Hospital, Aarhus, Denmark
| | - Johan Lindberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Karina Dalsgaard Sørensen
- Department of Molecular Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus, 8200, Denmark.
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
12
|
Zhao Z, Jing Y, Xu Z, Zhao H, He X, Lu T, Bai J, Qin W, Yang L. The mechanism of histone modifications in regulating enzalutamide sensitivity in advanced prostate cancer. Int J Biol Sci 2025; 21:2880-2890. [PMID: 40303302 PMCID: PMC12035886 DOI: 10.7150/ijbs.109638] [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: 12/30/2024] [Accepted: 03/23/2025] [Indexed: 05/02/2025] Open
Abstract
Prostate cancer (PCa) is the second most common malignant tumor in men worldwide, particularly castration-resistant prostate cancer (CRPC), for which enzalutamide (Enz) resistance is of particular concern. Modifications to histone methylation and acetylation patterns are closely associated with resistance to Enz in these patients. As PCa progresses, cancer cells alter their histone modification patterns, leading to a reduction in Enz treatment efficacy. Signaling pathways in the tumor microenvironment regulate gene expression by affecting the activity of histone-modifying enzymes, further affecting the efficacy of Enz. This review summarizes recent research about changes in histone modification patterns that occur in drug resistance-related genes at different stages of PCa and explores the potential use of combination therapies for reversing this process, providing insights into novel treatment strategies to improve the clinical efficacy of Enz.
Collapse
Affiliation(s)
- Zhite Zhao
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Yuming Jing
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Zhicheng Xu
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Hongfan Zhao
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xinglin He
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
- The Second Clinical Medical College, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi 710000, China
| | - Tong Lu
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jianhui Bai
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
- Department of Urology, Joint Logistics Support Force, Hospital 987, Baoji, China
| | - Weijun Qin
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Lijun Yang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| |
Collapse
|
13
|
Bergmann L, Greimeier S, Riethdorf S, Rohlfing T, Kaune M, Busenbender T, Strewinsky N, Dyshlovoy S, Joosse S, Peine S, Pantel K, von Amsberg G, Werner S. Transcriptional profiles of circulating tumor cells reflect heterogeneity and treatment resistance in advanced prostate cancer. J Exp Clin Cancer Res 2025; 44:111. [PMID: 40181402 PMCID: PMC11967125 DOI: 10.1186/s13046-025-03367-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/17/2025] [Indexed: 04/05/2025] Open
Abstract
PURPOSE New biomarkers for the detection and monitoring of aggressive variant prostate cancer (AVPC) including therapy-induced neuroendocrine prostate cancer (NEPC) are urgently needed, as measuring prostate-specific antigen (PSA) is not reliable in androgen-indifferent diseases. Molecular analysis of circulating tumor cells (CTC) enables repeated analysis for monitoring and allows to capture the heterogeneity of the disease. EXPERIMENTAL DESIGN 102 blood samples from 76 metastatic prostate cancer (mPC) patients, including 37 samples from histologically proven NEPC, were collected and CTCs were enriched using label-dependent and label-independent methods. Relevant transcripts were selected for CTC profiling using semi-quantitative RT-PCR analysis and validated in published datasets and cell lines. Transcriptional profiles in patient samples were analyzed using supervised and unsupervised methods. RESULTS CTC counts were increased in AVPC and NEPC as compared to metastatic hormone-sensitive prostate cancer (mHSPC). Gene expression profiles of CTCs showed a high degree of inter-patient heterogeneity, but NEPC-specific transcripts were significantly increased in patients with proven NEPC, while adenocarcinoma markers were decreased. Unsupervised analysis identified four distinct clusters of CTClow, ARhigh, amphicrine and pure NEPC gene expression profiles that reflected the clinical groups. Based on the transcript panel, NEPC could be distinguished from mHSPC or AVPC patients with a specificity of 95.5% and 88.2%, respectively. CONCLUSION Molecular subtypes of mPC can be distinguished by transcriptional profiling of CTCs. In the future, our convenient PCR-based analysis may complement the monitoring of advanced PCa patients and allow timely detection of resistance to androgen receptor pathway inhibitors.
Collapse
Affiliation(s)
- Lina Bergmann
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Sarah Greimeier
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Sabine Riethdorf
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Tina Rohlfing
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
- Department of Hematology and Oncology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Moritz Kaune
- Martini-Klinik, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Tobias Busenbender
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
- Department of Hematology and Oncology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Nadja Strewinsky
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
- Department of Hematology and Oncology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Sergey Dyshlovoy
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
- Department of Hematology and Oncology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Simon Joosse
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
- Mildred Scheel Cancer Career Centre HaTriCS4, University Medical Centre Hamburg- Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Sven Peine
- Department of Transfusion Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Klaus Pantel
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Gunhild von Amsberg
- European Liquid Biopsy Society (ELBS), Hamburg, Germany.
- Department of Hematology and Oncology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
- Martini-Klinik, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
| | - Stefan Werner
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
- European Liquid Biopsy Society (ELBS), Hamburg, Germany.
- Mildred Scheel Cancer Career Centre HaTriCS4, University Medical Centre Hamburg- Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
| |
Collapse
|
14
|
Wang H, Zhang S, Pan Q, Guo J, Li N, Chen L, Xu J, Zhou J, Gu Y, Wang X, Zhang G, Lian Y, Zhang W, Lin N, Jin Z, Zang Y, Lan W, Cheng X, Tan M, Chen FX, Jiang J, Liu Q, Zheng M, Qin J. Targeting the histone reader ZMYND8 inhibits antiandrogen-induced neuroendocrine tumor transdifferentiation of prostate cancer. NATURE CANCER 2025; 6:629-646. [PMID: 40102673 DOI: 10.1038/s43018-025-00928-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 02/10/2025] [Indexed: 03/20/2025]
Abstract
The transdifferentiation from adenocarcinoma to neuroendocrine prostate cancer (NEPC) in men confers antiandrogen therapy resistance. Here our analysis combining CRISPR‒Cas9 screening with single-cell RNA sequencing tracking of tumor transition demonstrated that antiandrogen-induced zinc finger MYND-type containing 8 (ZMYND8)-dependent epigenetic programming orchestrates NEPC transdifferentiation. Ablation of Zmynd8 prevents NEPC development, while ZMYND8 upregulation mediated by achaete-scute homolog 1 promotes NEPC differentiation. We show that forkhead box protein M1 (FOXM1) stabilizes ZMYND8 binding to chromatin regions characterized by H3K4me1-H3K14ac modification and FOXM1 targeting. Antiandrogen therapy releases the SWI/SNF chromatin remodeling complex from the androgen receptor, facilitating its interaction with ZMYND8-FOXM1 to upregulate critical neuroendocrine lineage regulators. We develop iZMYND8-34, a small molecule designed to inhibit ZMYND8's histone recognition, which effectively blocks NEPC development. These findings reveal the critical role of ZMYND8-dependent epigenetic programming induced by androgen deprivation therapy in orchestrating lineage fate. Targeting ZMYND8 emerges as a promising strategy for impeding NEPC development.
Collapse
Affiliation(s)
- Hanling Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Sulin Zhang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Qiang Pan
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
- Jinfeng Laboratory, Chongqing, China
| | - Jiacheng Guo
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Ni Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
- Jinfeng Laboratory, Chongqing, China
| | - Lifan Chen
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Junyu Xu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Jingyi Zhou
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yongqiang Gu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Xuege Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Guoying Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Yannan Lian
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Wei Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Naiheng Lin
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Zige Jin
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Yi Zang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Weihua Lan
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, China
| | | | - Minjia Tan
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Fei Xavier Chen
- Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jun Jiang
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, China
| | - Qiuli Liu
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, China.
| | - Mingyue Zheng
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.
| | - Jun Qin
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China.
- Jinfeng Laboratory, Chongqing, China.
| |
Collapse
|
15
|
Séguier D, Parent P, Duterque-Coquillaud M, Labreuche J, Fromont-Hankard G, Dariane C, Penel N, Villers A, Turpin A, Olivier J. Emergence of Neuroendocrine Tumors in Patients Treated with Androgen Receptor Pathway Inhibitors for Metastatic Prostate Cancer: A Systematic Review and Meta-analysis. Eur Urol Oncol 2025; 8:581-590. [PMID: 39824723 DOI: 10.1016/j.euo.2024.12.014] [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: 09/24/2024] [Revised: 11/28/2024] [Accepted: 12/24/2024] [Indexed: 01/20/2025]
Abstract
BACKGROUND AND OBJECTIVE It has been shown that androgen receptor pathway inhibitor (ARPIs) treatment for metastatic castration-resistant prostate cancer (mCRPC) improves overall survival rates, but ARPIs appear to be associated with a higher frequency of treatment-related neuroendocrine prostate cancer (t-NEPC). Our aim was to quantify the proportion of prostate adenocarcinoma cases that transition to t-NEPC following ARPI therapy. METHODS We conducted a comprehensive search of the literature on t-NEPC using databases including MEDLINE and Scopus. Eligible studies reported outcome data for NEPC in patients with prior mCRPC treated with an ARPI. To determine the pooled frequency of neuroendocrine transformation, the Freeman-Tukey variance-stabilizing arcsine transformation was applied to individual frequencies. KEY FINDINGS AND LIMITATIONS Among the 938 patients in eight eligible studies, t-NEPC diagnosis was confirmed in 171 patients, predominantly via pathology. Baseline biopsy verification to ensure the absence of NEPC was performed in most cases. The definition of t-NEPC varied among the studies. Five studies used a morphological definition based on histopathology, and three studies used NEPC biomarker detection on circulating tumor cells. A meta-analysis of aggregate data revealed an overall NEPC frequency following ARPI therapy of 16% (95% confidence interval 9-24%). CONCLUSION AND CLINICAL IMPLICATIONS ARPI-related NEPC represents a frequently underdiagnosed late complication of mCRPC. Given the absence of biomarkers for diagnosis, routine repeat biopsy at the mCRPC stage should be considered to diagnose t-NEPC transitions.
Collapse
Affiliation(s)
- Denis Séguier
- Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER) Research Group, CHU Lille, Institut Pasteur de Lille, and University of Lille, Lille, France; Department of Urology, Hospital Claude Huriez, CHU Lille, Lille, France
| | - Pauline Parent
- Department of Medical Oncology, CHU Lille, Lille, France
| | - Martine Duterque-Coquillaud
- Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER) Research Group, CHU Lille, Institut Pasteur de Lille, and University of Lille, Lille, France
| | | | | | - Charles Dariane
- Service d'Urologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France; Université Paris Cité, Paris, France
| | - Nicolas Penel
- Department of Medical Oncology, Centre Oscar Lambret, University of Lille, Lille, France
| | - Arnauld Villers
- Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER) Research Group, CHU Lille, Institut Pasteur de Lille, and University of Lille, Lille, France; Department of Urology, Hospital Claude Huriez, CHU Lille, Lille, France
| | - Anthony Turpin
- Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER) Research Group, CHU Lille, Institut Pasteur de Lille, and University of Lille, Lille, France; Department of Medical Oncology, CHU Lille, Lille, France
| | - Jonathan Olivier
- Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER) Research Group, CHU Lille, Institut Pasteur de Lille, and University of Lille, Lille, France; Department of Urology, Hospital Claude Huriez, CHU Lille, Lille, France.
| |
Collapse
|
16
|
Zhu X, Ding CKC, Aggarwal RR. Emerging Therapeutic Targets of Neuroendocrine Prostate Cancer. Curr Oncol Rep 2025; 27:362-374. [PMID: 40011325 DOI: 10.1007/s11912-025-01643-9] [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] [Accepted: 01/27/2025] [Indexed: 02/28/2025]
Abstract
PURPOSE OF REVIEW Treatment-emergent neuroendocrine prostate cancer (NEPC) is aggressive and lethal. As androgen receptor signaling inhibitors (ARSIs) are increasingly used in earlier disease settings, treatment-emergent NEPC becomes more prevalent, and effective therapies are urgently needed. The purpose of this review was to summarize recent progress on emerging therapeutic targets of NEPC. RECENT FINDINGS A multitude of therapeutic targets have emerged in NEPC over recent years. These targets may represent drivers of treatment-emergent lineage plasticity or simply be overexpressed on the surface of NEPC cells. Multiple modalities have been employed to drug these targets, with promising preclinical and clinical results. Treatment-emergent NEPC represents a distinct and clinically significant subset of castration-resistant prostate cancer (CRPC). Emerging therapeutic approaches have demonstrated encouraging efficacy and safety profiles, offering the potential to improve patient outcomes.
Collapse
Affiliation(s)
- Xiaolin Zhu
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Chien-Kuang C Ding
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Department of Anatomic Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Rahul R Aggarwal
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
17
|
Dunmore KE, Rickman DS. Targeting anti-androgen therapy resistance through epigenetic rewiring. NATURE CANCER 2025; 6:564-566. [PMID: 40102672 DOI: 10.1038/s43018-025-00906-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Affiliation(s)
- Kate E Dunmore
- Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - David S Rickman
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA.
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
18
|
Kang N, Xue H, Wong NK, Lin YY, Classen A, Wu R, Oo HZ, Dong X, Trinh A, Lin D, Daugaard M, Ong C, Collins C, Gleave M, Wang Y. Exploring B7-H4's Role in Prostate Cancer Dormancy after Androgen Deprivation Therapy: Extracellular Matrix Interactions and Therapeutic Opportunities. Mol Cancer Res 2025; 23:327-338. [PMID: 39776163 PMCID: PMC11972443 DOI: 10.1158/1541-7786.mcr-24-0958] [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: 10/11/2024] [Revised: 12/13/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025]
Abstract
Prostate cancer is mainly managed with androgen deprivation therapy (ADT), but this often leads to a dormant state and subsequent relapse as lethal castration-resistant prostate cancer (CRPC). Using our unique prostate cancer patient-derived xenograft dormancy models, we investigated this critical dormant phase and discovered a selective increase in B7-H4 expression during the dormancy period following mouse host castration. This finding is supported by observations in clinical specimens of patients with prostate cancer treated with ADT. Differential expression analyses revealed the enrichment of extracellular matrix (ECM)-cell interaction pathways in B7-H4-positive cells. Functional assays demonstrated a crucial role of B7-H4 in maintaining dormancy within the ECM niche. Specifically, B7-H4 expression in LNCaP cells reduced proliferation within the dormant ECM in vitro and significantly delayed relapse in castrated hosts in vivo. These results shed light on the dynamic regulation of B7-H4 during prostate cancer dormancy and underscore its potential as a therapeutic target for preventing CRPC relapse. Implications: Our study identified membranous B7-H4 expression during ADT-induced dormancy, highlighting its potential as a therapeutic target for managing dormant prostate cancer and preventing fatal CRPC relapse.
Collapse
Affiliation(s)
- Ning Kang
- Department of Experimental Therapeutics, BC Cancer, Vancouver, BC, Canada
| | - Hui Xue
- Department of Experimental Therapeutics, BC Cancer, Vancouver, BC, Canada
| | - Nelson K.Y. Wong
- Department of Experimental Therapeutics, BC Cancer, Vancouver, BC, Canada
| | - Yen-Yi Lin
- Vancouver Prostate Centre, Vancouver, BC, Canada
| | - Adam Classen
- Department of Experimental Therapeutics, BC Cancer, Vancouver, BC, Canada
- Vancouver Prostate Centre, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Rebecca Wu
- Department of Experimental Therapeutics, BC Cancer, Vancouver, BC, Canada
| | - Htoo Zarni Oo
- Vancouver Prostate Centre, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Xin Dong
- Department of Experimental Therapeutics, BC Cancer, Vancouver, BC, Canada
| | | | - Dong Lin
- Department of Experimental Therapeutics, BC Cancer, Vancouver, BC, Canada
- Vancouver Prostate Centre, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Mads Daugaard
- Vancouver Prostate Centre, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Christopher Ong
- Vancouver Prostate Centre, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Colin Collins
- Vancouver Prostate Centre, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Martin Gleave
- Vancouver Prostate Centre, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yuzhuo Wang
- Department of Experimental Therapeutics, BC Cancer, Vancouver, BC, Canada
- Vancouver Prostate Centre, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
19
|
Woytash JA, Kumar R, Chaudhary AK, Donnelly C, Wojtulski A, Bethu M, Wang J, Spernyak J, Bross P, Yadav N, Inigo JR, Chandra D. Mitochondrial unfolded protein response-dependent β-catenin signaling promotes neuroendocrine prostate cancer. Oncogene 2025; 44:820-834. [PMID: 39690273 DOI: 10.1038/s41388-024-03261-4] [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: 06/19/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 12/19/2024]
Abstract
The mitochondrial unfolded protein response (UPRmt) maintains mitochondrial quality control and proteostasis under stress conditions. However, the role of UPRmt in aggressive and resistant prostate cancer is not clearly defined. We show that castration-resistant neuroendocrine prostate cancer (CRPC-NE) harbored highly dysfunctional oxidative phosphorylation (OXPHOS) Complexes. However, biochemical and protein analyses of CRPC-NE tumors showed upregulation of nuclear-encoded OXPHOS proteins and UPRmt in this lethal subset of prostate cancer suggestive of compensatory upregulation of stress signaling. Genetic deletion and pharmacological inhibition of the main chaperone of UPRmt heat shock protein 60 (HSP60) reduced neuroendocrine prostate cancer (NEPC) growth in vivo as well as reverted NEPC cells to a more epithelial-like state. HSP60-dependent aggressive NEPC phenotypes was associated with upregulation of β-catenin signaling both in cancer cells and in vivo tumors. HSP60 expression rendered enrichment of aggressive prostate cancer signatures and metastatic potential were inhibited upon suppression of UPRmt. We discovered that UPRmt promoted OXPHOS functions including mitochondrial bioenergetics in CRPC-NE via regulation of β-catenin signaling. Mitochondrial biogenesis facilitated cisplatin resistance and inhibition of UPRmt resensitizes CRPC-NE cells to cisplatin. Together, our findings demonstrated that UPRmt promotes mitochondrial health via upregulating β-catenin signaling and UPRmt represents viable therapeutic target for NEPC.
Collapse
Affiliation(s)
- Jordan Alyse Woytash
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Rahul Kumar
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Ajay K Chaudhary
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Cullan Donnelly
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Adam Wojtulski
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Murali Bethu
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Jianmin Wang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Joseph Spernyak
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Peter Bross
- Research Unit for Molecular Medicine, Aarhus University and Aarhus University Hospital, 8200, Aarhus N, Denmark
| | - Neelu Yadav
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Joseph R Inigo
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Dhyan Chandra
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
| |
Collapse
|
20
|
Feng EM, Vo-Phamhi J, Subramanian AN, Dias M, Foye A, Vinson J, Hong JC, Freedland SJ, Alumkal JJ, Beltran H, Morrissey C, Nelson PS, Chinnaiyan AM, Aggarwal R, Small EJ, Quigley DA, Sjöström M, Zhao SG, Chen WS. Racial variation in the advanced prostate cancer genome. Prostate Cancer Prostatic Dis 2025:10.1038/s41391-025-00949-w. [PMID: 40164700 DOI: 10.1038/s41391-025-00949-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/10/2024] [Accepted: 02/07/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Racial differences in metastatic castration-resistant prostate cancer (mCRPC) genomes have not yet been fully studied. We aimed to investigate transcriptomic, mutational, and clinical differences by race in a large multi-institutional cohort of men with mCRPC. METHODS Genomic and clinicopathologic data from four mCRPC tumor biopsy cohorts were obtained and aggregated. Gene set enrichment analyses were performed to assess pathway-level differences in gene expression by patient race. DNA alteration frequencies of known prostate cancer driver genes and clinical outcomes were compared across racial groups. RESULTS In our cohort of 445 men with mCRPC, tumors from African American patients (N = 26) demonstrated higher expression of MYC pathway genes (FDR q = 0.03) and lower expression of IFN-γ, IL-6/JAK/STAT3, and inflammatory pathway genes (FDR q < 0.001) compared to tumors from European American patients. TMPRSS2:ERG gene fusions were observed more frequently in tumors from European American compared to African American patients (41% vs. 11%, P = 0.015). Asian patients (N = 9) and other racial groups comprised a small minority of our cohort. No differences in overall survival were noted across racial groups. CONCLUSIONS Despite demonstrating similar clinical outcomes, cancers from African Americans display distinct tumor biology. Specifically, we observed racial differences in expression of prostate cancer driver gene pathways (including potential clinically actionable pathways of IFN-γ and JAK/STAT) and DNA alterations, including TMPRSS2:ERG gene fusion. Our findings highlight the importance of racial diversity in future genomic profiling and clinical trials efforts.
Collapse
Affiliation(s)
- Emily M Feng
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Jenny Vo-Phamhi
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Aishwarya N Subramanian
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Mikhail Dias
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Adam Foye
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Jake Vinson
- Prostate Cancer Clinical Trials Consortium (PCCTC), New York, NY, USA
| | - Julian C Hong
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Stephen J Freedland
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Section of Urology, Durham VA Medical Center, Durham, NC, USA
| | - Joshi J Alumkal
- Department of Internal Medicine, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Colm Morrissey
- Department of Urology, University of Washington School of Medicine, Seattle, WA, USA
| | - Peter S Nelson
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Arul M Chinnaiyan
- Departments of Pathology and Urology, University of Michigan, Ann Arbor, MI, USA
| | - Rahul Aggarwal
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Eric J Small
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - David A Quigley
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Department of Urology, University of California San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Martin Sjöström
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Department of Clinical Sciences Lund, Division of Oncology, Lund University, Lund, Sweden
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Shuang G Zhao
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - William S Chen
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
21
|
Guzmán-Esquivel J, Garcia-Garcia HS, Hernández-Fuentes GA, Venegas-Ramírez J, Barajas-Mejía CD, Garza-Veloz I, Martinez-Fierro ML, Magaña-Vergara NE, Guzmán-Solórzano JA, Calvo-Soto P, Avila-Zamora ON, Fuentes-Murguia M, Ceja-Espíritu G, Delgado-Enciso I. The Impact of Doxycycline as an Adjunctive Therapy on Prostate-Specific Antigen, Quality of Life, and Cognitive Function in Metastatic Prostate Cancer Patients: A Phase II Randomized Controlled Trial. Pharmaceutics 2025; 17:404. [PMID: 40284400 PMCID: PMC12030710 DOI: 10.3390/pharmaceutics17040404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 03/13/2025] [Accepted: 03/16/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Metastatic prostate cancer remains a major clinical challenge, with limited therapeutic options. Doxycycline, a tetracycline antibiotic with anti-inflammatory properties, has shown potential as an adjunctive therapy. This study aimed to evaluate its efficacy in reducing prostate-specific antigen (PSA) levels and improving quality of life in patients receiving standard treatment for metastatic prostate cancer. Methods: This phase II, double-blind, randomized controlled trial included 45 participants (aged 57-81 years) assigned to doxycycline (100 mg daily) or a placebo for six months. The primary outcome was the percentage change in PSA levels at 3 and 6 months. Secondary outcomes included quality of life (EQ-5D-5L), cognitive function (Mini-Mental State Examination), and glucose levels. Additionally, a structure-activity relationship (SAR) analysis was performed through an extensive bibliographic review to identify pharmacophores responsible for doxycycline's biological activity, particularly its tetracyclic core. The SAR analysis included tetracyclines and derivatives, androgen-targeting agents, and other pharmacologically relevant molecules used in prostate cancer therapy. Statistical analysis was conducted using multivariate logistic regression. Results: At six months, the doxycycline group showed a median PSA reduction of 60% compared to 10% in the placebo group (p = 0.043). A ≥50% reduction in PSA levels was observed in 71.4% of patients receiving doxycycline versus 20.8% in the placebo group (p = 0.001), with an adjusted relative risk of 10.309 (95% CI: 2.359-45.055, p = 0.002). Quality of life improved, with 7.1% of doxycycline-treated patients reporting poor quality of life compared to 42.9% in the placebo group (p = 0.028). A slight improvement in cognitive function was also noted (p = 0.037). SAR analysis suggested that the tetracyclic ring of doxycycline may play a crucial role in its observed biological effects. Conclusions: Doxycycline demonstrates potential as an adjunctive therapy in metastatic prostate cancer by reducing PSA levels and improving quality of life. The SAR analysis supports the hypothesis that its tetracyclic structure may be responsible for its therapeutic effects. Further large-scale trials are warranted to confirm these findings.
Collapse
Affiliation(s)
- José Guzmán-Esquivel
- Clinical Epidemiology Research Unit, Mexican Institute of Social Security Institute, Villa de Alvarez, Colima 28984, Mexico; (J.G.-E.); (H.S.G.-G.); (C.D.B.-M.)
| | - Hossana S. Garcia-Garcia
- Clinical Epidemiology Research Unit, Mexican Institute of Social Security Institute, Villa de Alvarez, Colima 28984, Mexico; (J.G.-E.); (H.S.G.-G.); (C.D.B.-M.)
| | - Gustavo A. Hernández-Fuentes
- Department of Molecular Medicine, School of Medicine, University of Colima, Colima 28040, Mexico; (G.A.H.-F.); (J.A.G.-S.); (M.F.-M.); (G.C.-E.)
- State Cancerology Institute of Colima, Health Services of the Mexican Social Security Institute for Welfare (IMSS-BIENESTAR), Colima 28085, Mexico;
- Faculty of Chemical Sciences, University of Colima, Coquimatlan 28400, Mexico;
| | - Jesús Venegas-Ramírez
- Department of Nephrology, Mexican Institute of Social Security (IMSS), General Hospital of Zone No. 1, IMSS, Villa de Alvarez 28984, Mexico;
| | - Carlos D. Barajas-Mejía
- Clinical Epidemiology Research Unit, Mexican Institute of Social Security Institute, Villa de Alvarez, Colima 28984, Mexico; (J.G.-E.); (H.S.G.-G.); (C.D.B.-M.)
| | - Idalia Garza-Veloz
- Molecular Medicine Laboratory, Unidad de Medicina Humana y Ciencias de la Salud, Universidad Autónoma de Zacatecas, Zacatecas 98160, Mexico; (I.G.-V.); (M.L.M.-F.)
| | - Margarita L. Martinez-Fierro
- Molecular Medicine Laboratory, Unidad de Medicina Humana y Ciencias de la Salud, Universidad Autónoma de Zacatecas, Zacatecas 98160, Mexico; (I.G.-V.); (M.L.M.-F.)
| | - Nancy E. Magaña-Vergara
- Faculty of Chemical Sciences, University of Colima, Coquimatlan 28400, Mexico;
- Consejo Nacional de Humanidades, Ciencia y Tecnología (CONAHCYT), Mexico City 03940, Mexico
| | - José A. Guzmán-Solórzano
- Department of Molecular Medicine, School of Medicine, University of Colima, Colima 28040, Mexico; (G.A.H.-F.); (J.A.G.-S.); (M.F.-M.); (G.C.-E.)
| | - Patricia Calvo-Soto
- Coordination of Planning and Institutional Liaison, IMSS OOAD Colima, Colima 28030, Mexico;
| | - Oscar N. Avila-Zamora
- State Cancerology Institute of Colima, Health Services of the Mexican Social Security Institute for Welfare (IMSS-BIENESTAR), Colima 28085, Mexico;
| | - Mercedes Fuentes-Murguia
- Department of Molecular Medicine, School of Medicine, University of Colima, Colima 28040, Mexico; (G.A.H.-F.); (J.A.G.-S.); (M.F.-M.); (G.C.-E.)
| | - Gabriel Ceja-Espíritu
- Department of Molecular Medicine, School of Medicine, University of Colima, Colima 28040, Mexico; (G.A.H.-F.); (J.A.G.-S.); (M.F.-M.); (G.C.-E.)
| | - Iván Delgado-Enciso
- Department of Molecular Medicine, School of Medicine, University of Colima, Colima 28040, Mexico; (G.A.H.-F.); (J.A.G.-S.); (M.F.-M.); (G.C.-E.)
- State Cancerology Institute of Colima, Health Services of the Mexican Social Security Institute for Welfare (IMSS-BIENESTAR), Colima 28085, Mexico;
- Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
22
|
Meng Y, Ge J, Zhou C, Ma H, Chen C, Zhou Y, Hu X, Xu Y, Wang X, Shi G, Yu W, Zhang J. Elevated VRK1 levels after androgen deprivation therapy promote prostate cancer progression by upregulating YAP1 expression. J Cancer Res Clin Oncol 2025; 151:116. [PMID: 40111564 PMCID: PMC11926012 DOI: 10.1007/s00432-025-06168-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025]
Abstract
PURPOSE Vaccinia-related kinase 1 (VRK1) is a serine-threonine kinase involved in the proliferation and migration of various cancer cells. However, its role in prostate cancer (PCa), particularly in the development of therapeutic resistance, remains unclear. METHODS We established an androgen-independent PCa cell line derived from LNCaP prostate cancer cells and conducted transcriptome and proteome sequencing together with bioinformatic analyses of large clinical sample databases to investigate the potential role of VRK1 in PCa progression. The correlation between VRK1 and androgen receptor (AR) signaling was evaluated under simulated clinical treatment conditions. The effects of VRK1 on cell proliferation were assessed in vitro and in vivo using Cell Counting Kit-8 and colony formation assays. Additionally, proteome and transcriptome sequencing, combined with rescue experiments were performed to explore VRK1-regulated signaling pathways related to cell proliferation and therapeutic resistance. RESULTS VRK1 expression was elevated during the progression of androgen-dependent prostate cancer to castration-resistant prostate cancer under therapeutic conditions, and high VRK1 expression was associated with a poor prognosis in patients with PCa. VRK1 was regulated by AR signaling, and its silencing suppressed PCa cell proliferation both in vitro and in vivo. VRK1 drove cell proliferation and therapeutic resistance in PCa by modulating yes-associated protein 1 (YAP1). CONCLUSIONS VRK1 serves as a prognostic marker in PCa, regulated by AR signaling. VRK1 depletion inhibited cell proliferation both in vitro and in vivo, while elevated VRK1 upregulated YAP1, promoting cell proliferation and therapeutic resistance.
Collapse
MESH Headings
- Humans
- Male
- YAP-Signaling Proteins
- Disease Progression
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Adaptor Proteins, Signal Transducing/biosynthesis
- Animals
- Cell Proliferation
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Mice
- Protein Serine-Threonine Kinases/metabolism
- Protein Serine-Threonine Kinases/genetics
- Prostatic Neoplasms/pathology
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/drug therapy
- Up-Regulation
- Receptors, Androgen/metabolism
- Intracellular Signaling Peptides and Proteins/metabolism
- Intracellular Signaling Peptides and Proteins/genetics
- Gene Expression Regulation, Neoplastic
- Cell Line, Tumor
- Prostatic Neoplasms, Castration-Resistant/pathology
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/metabolism
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prognosis
- Mice, Nude
- Androgen Antagonists/pharmacology
- Signal Transduction
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Yibo Meng
- Department of Urology, The Fifth People'S Hospital of Shanghai, Fudan University, No. 801, Heqing Road, Minhang District, Shanghai, 200240, People's Republic of China
| | - Jianchao Ge
- Department of Urology, The Fifth People'S Hospital of Shanghai, Fudan University, No. 801, Heqing Road, Minhang District, Shanghai, 200240, People's Republic of China
| | - Cheng Zhou
- Department of Urology, The Fifth People'S Hospital of Shanghai, Fudan University, No. 801, Heqing Road, Minhang District, Shanghai, 200240, People's Republic of China
| | - Hangbin Ma
- Department of Urology, The Fifth People'S Hospital of Shanghai, Fudan University, No. 801, Heqing Road, Minhang District, Shanghai, 200240, People's Republic of China
| | - Chenchen Chen
- Department of Urology, The Fifth People'S Hospital of Shanghai, Fudan University, No. 801, Heqing Road, Minhang District, Shanghai, 200240, People's Republic of China
| | - Yinghao Zhou
- Department of Urology, The Fifth People'S Hospital of Shanghai, Fudan University, No. 801, Heqing Road, Minhang District, Shanghai, 200240, People's Republic of China
| | - Xuetao Hu
- Department of Urology, The Fifth People'S Hospital of Shanghai, Fudan University, No. 801, Heqing Road, Minhang District, Shanghai, 200240, People's Republic of China
| | - Yaozong Xu
- Department of Urology, The Fifth People'S Hospital of Shanghai, Fudan University, No. 801, Heqing Road, Minhang District, Shanghai, 200240, People's Republic of China
| | - Xilong Wang
- Department of Urology, The Fifth People'S Hospital of Shanghai, Fudan University, No. 801, Heqing Road, Minhang District, Shanghai, 200240, People's Republic of China
| | - Guowei Shi
- Department of Urology, The Fifth People'S Hospital of Shanghai, Fudan University, No. 801, Heqing Road, Minhang District, Shanghai, 200240, People's Republic of China.
| | - Wandong Yu
- Department of Urology, The Fifth People'S Hospital of Shanghai, Fudan University, No. 801, Heqing Road, Minhang District, Shanghai, 200240, People's Republic of China.
| | - Jun Zhang
- Department of Urology, The Fifth People'S Hospital of Shanghai, Fudan University, No. 801, Heqing Road, Minhang District, Shanghai, 200240, People's Republic of China.
| |
Collapse
|
23
|
Zhang T, Wang B, Wei Y, Gan H, Fang B, Li X, Wu J, Bian X, Wang J, Freedland SJ, Huang S, Ye D, Zhu Y. Neoadjuvant fuzuloparib combined with abiraterone for localized high-risk prostate cancer (FAST-PC): A single-arm phase 2 study. Cell Rep Med 2025; 6:102018. [PMID: 40056906 PMCID: PMC11970390 DOI: 10.1016/j.xcrm.2025.102018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/19/2024] [Accepted: 02/17/2025] [Indexed: 03/21/2025]
Abstract
Preclinical studies suggest synergistic effects between androgen receptor inhibitors and poly(adenosine diphosphate-ribose) polymerase (PARP) inhibitors. This phase 2 trial (NCT05223582) evaluates neoadjuvant fuzuloparib plus abiraterone in 35 treatment-naive men with localized high-risk prostate cancer. Patients receive six cycles of therapy followed by radical prostatectomy. Primary endpoints are pathological complete response (pCR) and minimal residual disease (MRD, ≤5 mm). The combined pCR/MRD rate is 46% (95% confidence interval [CI]: 29%-63%), with a 53% 2-year biochemical progression-free survival rate. Grade ≥3 adverse events occur in 23% of patients. Biallelic homologous recombination repair/BRCA2 alterations correlate with faster prostate-specific antigen decline. Post-treatment genomic analyses reveal reduced MYC amplification and proliferation markers, alongside activated epithelial-mesenchymal transition/activator protein 1 (AP-1) pathways. The trial meets its primary endpoint, demonstrating feasibility and preliminary efficacy, while exploratory biomarkers may guide future studies.
Collapse
Affiliation(s)
- Tingwei Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Beihe Wang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Yu Wei
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Hualei Gan
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Bangwei Fang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Xiaomeng Li
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Junlong Wu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Xiaojie Bian
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Jianfei Wang
- Jiangsu Hengrui Pharmaceuticals Co., Ltd, Shanghai, China
| | - Stephen J Freedland
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Section of Urology, Durham VA Medical Center, Durham, NC, USA
| | - Shenglin Huang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Department of Medical Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Genitourinary Cancer Institute, Shanghai, China.
| | - Yao Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Genitourinary Cancer Institute, Shanghai, China.
| |
Collapse
|
24
|
Li S, Song K, Sun H, Tao Y, Huang A, Bhatia V, Hanratty B, Patel RA, Long HW, Morrissey C, Haffner MC, Nelson PS, Graeber TG, Lee JK. Defined cellular reprogramming of androgen receptor-active prostate cancer to neuroendocrine prostate cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637904. [PMID: 40027790 PMCID: PMC11870442 DOI: 10.1101/2025.02.12.637904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Neuroendocrine prostate cancer (NEPC) arises primarily through neuroendocrine transdifferentiation (NEtD) as an adaptive mechanism of therapeutic resistance. Models to define the functional effects of putative drivers of this process on androgen receptor (AR) signaling and NE cancer lineage programs are lacking. We adapted a genetically defined strategy from the field of cellular reprogramming to directly convert AR-active prostate cancer (ARPC) to AR-independent NEPC using candidate factors. We delineated critical roles of the pioneer factors ASCL1 and NeuroD1 in NEtD and uncovered their abilities to silence AR expression and signaling by remodeling chromatin at the somatically acquired AR enhancer and global AR binding sites with enhancer activity. We also elucidated the dynamic temporal changes in the transcriptomic and epigenomic landscapes of cells undergoing acute lineage conversion from ARPC to NEPC which should inform future therapeutic development. Further, we distinguished the activities of ASCL1 and NeuroD1 from the inactivation of RE-1 silencing transcription factor (REST), a master suppressor of a major neuronal gene program, in establishing a NEPC lineage state and in modulating the expression of genes associated with major histocompatibility complex class I (MHC I) antigen processing and presentation. These findings provide important, clinically relevant insights into the biological processes driving NEtD of prostate cancer.
Collapse
Affiliation(s)
- Shan Li
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Kai Song
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Huiyun Sun
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, 98195, USA
| | - Yong Tao
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Arthur Huang
- Department of Urology, University of Washington, Seattle, WA, 98195, USA
| | - Vipul Bhatia
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Brian Hanratty
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Radhika A Patel
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Henry W Long
- Department of Medical Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
- Center for Functional Epigenetics, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, WA, 98195, USA
| | - Michael C Haffner
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
- Department of Pathology and Laboratory Medicine, University of Washington, Seattle, WA. 98195, USA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Peter S Nelson
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
- Department of Pathology and Laboratory Medicine, University of Washington, Seattle, WA. 98195, USA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Division of Medical Oncology, University of Washington, Seattle, WA, 98195, USA
| | - Thomas G Graeber
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
- Crump Institute for Molecular Imaging, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - John K Lee
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
- Institute of Urologic Oncology, Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| |
Collapse
|
25
|
Suzuki H, Akamatsu S, Shiota M, Kakiuchi H, Kimura T. Triplet therapy for metastatic castration-sensitive prostate cancer: Rationale and clinical evidence. Int J Urol 2025; 32:239-250. [PMID: 39651632 PMCID: PMC11923528 DOI: 10.1111/iju.15647] [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: 08/16/2024] [Accepted: 11/21/2024] [Indexed: 12/11/2024]
Abstract
Prostate cancer (PC) growth is hormone-dependent and it frequently develops distant metastases as disease progresses. Patients with metastatic castration-sensitive prostate cancer (mCSPC) initially respond to androgen deprivation therapy (ADT) but eventually become refractory and develop metastatic castration-resistant prostate cancer (mCRPC). Castration-resistance is associated with high lethality and metastases confer poor prognosis, therefore unmet needs in treatment for mCSPC remain high. So far, improvements in survival in mCSPC have been achieved by doublet combination therapy such as docetaxel or an androgen-receptor signaling inhibitor (ARSI) in addition to ADT. Further, recent phase 3 trials have shown that triplet therapy-a combination of ARSI, docetaxel, and ADT improves prognosis compared with docetaxel plus ADT in mCSPC. PC tumors manifest intra- and inter-tumoral heterogeneity at both the genetic and phenotypic level. As heterogeneity increases during sequential treatment and disease progression, it is reasonable to initiate combination therapy using drugs with different mechanisms of action early in the course of disease, such as mCSPC. Previous research about tumor heterogeneity and drug resistant mechanism support this rationale, as well as preclinical studies and real-world data provide the scientific evidence of benefit by combining ARSI and docetaxel. Here, we review the rationale and clinical evidence for triplet therapy in patients with mCSPC.
Collapse
Affiliation(s)
- Hiroyoshi Suzuki
- Department of UrologyToho University Sakura Medical CenterChibaJapan
| | | | | | - Haruka Kakiuchi
- Oncology Medical Affairs, Medical Affairs and PharmacovigilanceBayer Yakuhin Ltd.OsakaJapan
| | - Takahiro Kimura
- Department of UrologyThe Jikei University School of MedicineTokyoJapan
| |
Collapse
|
26
|
Fanelli GN, Nuzzo PV, Pederzoli F, Loda M. Deciphering Complexity: The Molecular Landscape of Castration-Resistant Prostate Cancer. Surg Pathol Clin 2025; 18:25-39. [PMID: 39890307 PMCID: PMC11787547 DOI: 10.1016/j.path.2024.10.003] [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: 02/03/2025]
Abstract
Despite improvements in diagnosis and treatment approaches, prostate cancer (PC) remains a leading cause of cancer-related death in men. PC progresses through various stages, mostly driven by androgen receptor signaling. However, after androgen deprivation therapies, in a significant portion of patients, several different molecular mechanisms contribute to the development of castration resistance. Delving deeply into the molecular landscape of castration-resistant PC, grasping the selective pressures exerted by therapies, and understanding the drivers of lineage plasticity is pivotal to prevent progression. Targeting genetic and epigenetic alterations that drive this transition will guide clinical management strategies and possibly prevent and/or treat lethal disease.
Collapse
Affiliation(s)
- Giuseppe Nicolò Fanelli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA; Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 57, Pisa 56125, Italy
| | - Pier Vitale Nuzzo
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA
| | - Filippo Pederzoli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
27
|
Sharifi MN, Feng E, Rydzewski NR, Taylor AK, Sperger JM, Shi Y, Helzer KT, Bootsma ML, Carreno V, Chang AH, Nunamaker LA, Blitzer GC, Shang TA, Subramanian A, Bjartell A, Josefsson A, Wikström P, Feng E, Kohli M, Yang R, Dehm SM, Small EJ, Aggarwal R, Quigley DA, Lang JM, Zhao SG, Sjöström M. Adverse prognosis gene expression patterns in metastatic castration-resistant prostate cancer. Mol Oncol 2025. [PMID: 39985777 DOI: 10.1002/1878-0261.70001] [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: 07/04/2024] [Revised: 11/21/2024] [Accepted: 02/10/2025] [Indexed: 02/24/2025] Open
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) is a heterogeneous disease. Several studies have identified transcriptional subtypes of mCRPC, but comprehensive analysis of prognostic gene expression pathways has been limited. Therefore, we aggregated a cohort of 1012 mCRPC tissue samples from 769 patients and investigated the association of gene expression-based pathways with clinical outcomes and intrapatient and intratumor heterogeneity. Survival data were obtained for 272 patients. Pathway-level enrichment was evaluated using gene set variation analysis. scRNA-seq datasets from mCRPC tissue biopsies and circulating tumor cells were used to investigate heterogeneity of adverse pathways. We identified five pathway clusters: (a) Immune response/WNT/TGF-beta signaling, (b) AR signaling/luminal signatures, (c) mTOR signaling and glycolysis, (d) cell proliferation, and (e) neuroendocrine differentiation. Proliferation, AR signaling loss, and glycolysis/mTOR signaling were independently prognostic. Adverse prognostic pathway scores decreased on treatment with AR signaling inhibitors, but not at progression, suggesting failure to permanently target these pathways. scRNA-seq datasets from mCRPC tissue biopsies and circulating tumor cells were used to investigate heterogeneity of adverse pathways. Our results suggest loss of AR signaling, high proliferation, and a glycolytic phenotype as adverse prognostic pathways in mCRPC that could be used in conjunction with clinical factors to prognosticate for treatment decisions.
Collapse
Affiliation(s)
- Marina N Sharifi
- Carbone Cancer Center, University of Wisconsin-Madison, WI, USA
- Department of Medicine, University of Wisconsin-Madison, WI, USA
| | - Eric Feng
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
- Department of Radiation Oncology, University of California, San Francisco, CA, USA
| | | | - Amy K Taylor
- Carbone Cancer Center, University of Wisconsin-Madison, WI, USA
- Department of Medicine, University of Wisconsin-Madison, WI, USA
| | - Jamie M Sperger
- Carbone Cancer Center, University of Wisconsin-Madison, WI, USA
- Department of Medicine, University of Wisconsin-Madison, WI, USA
| | - Yue Shi
- Department of Human Oncology, University of Wisconsin-Madison, WI, USA
| | - Kyle T Helzer
- Department of Human Oncology, University of Wisconsin-Madison, WI, USA
| | - Matthew L Bootsma
- Department of Human Oncology, University of Wisconsin-Madison, WI, USA
| | | | - Alex H Chang
- Department of Medicine, University of Wisconsin-Madison, WI, USA
| | | | - Grace C Blitzer
- Department of Human Oncology, University of Wisconsin-Madison, WI, USA
| | - Tianfu Andy Shang
- Department of Human Oncology, University of Wisconsin-Madison, WI, USA
| | - Aishwarya Subramanian
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
- Department of Radiation Oncology, University of California, San Francisco, CA, USA
| | - Anders Bjartell
- Department of Translational Medicine, Lund University, Malmö, Sweden
- Department of Urology, Skåne University Hospital, Malmö, Sweden
| | - Andreas Josefsson
- Department of Diagnostics and Interventions, Urology, Umeå University, Sweden
- Wallenberg Center for Molecular Medicine, Umeå University, Sweden
| | - Pernilla Wikström
- Department of Medical Biosciences, Pathology, Umeå University, Sweden
| | - Emily Feng
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
- Department of Radiation Oncology, University of California, San Francisco, CA, USA
| | - Manish Kohli
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Rendong Yang
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Scott M Dehm
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
- Department of Urology, University of Minnesota, Minneapolis, MN, USA
| | - Eric J Small
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, CA, USA
| | - Rahul Aggarwal
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, CA, USA
| | - David A Quigley
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Joshua M Lang
- Carbone Cancer Center, University of Wisconsin-Madison, WI, USA
- Department of Medicine, University of Wisconsin-Madison, WI, USA
| | - Shuang G Zhao
- Carbone Cancer Center, University of Wisconsin-Madison, WI, USA
- Department of Human Oncology, University of Wisconsin-Madison, WI, USA
- William S. Middleton Memorial Veterans' Hospital, Madison, WI, USA
| | - Martin Sjöström
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
- Department of Radiation Oncology, University of California, San Francisco, CA, USA
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Sweden
- Department of Hematology, Oncology, and Radiation Physics, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
28
|
Pitzen SP, Rudenick AN, Qiu Y, Zhang W, Munro SA, McCluskey BM, Forster C, Bergom HE, Ali A, Boytim E, Lafin JT, Linder S, Ismail M, Devlies W, Sessions CJ, Claessens F, Joniau S, Attard G, Zwart W, Nelson PS, Corey E, Wang Y, Lang JM, Beltran H, Strand D, Antonarakis ES, Hwang J, Murugan P, Huang RS, Dehm SM. Comparative transcriptomics reveals a mixed basal, club, and hillock epithelial cell identity in castration-resistant prostate cancer. Proc Natl Acad Sci U S A 2025; 122:e2415308122. [PMID: 39913208 PMCID: PMC11831193 DOI: 10.1073/pnas.2415308122] [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: 07/30/2024] [Accepted: 01/06/2025] [Indexed: 02/19/2025] Open
Abstract
Inhibiting the androgen receptor (AR) is effective for treatment of advanced prostate cancers because of their AR-dependent luminal epithelial cell identity. Tumors progress during therapy to castration-resistant prostate cancer (CRPC) by restoring AR signaling and maintaining luminal identity or by converting through lineage plasticity to a neuroendocrine (NE) identity or double-negative CRPC (DNPC) lacking luminal or NE identities. Here, we show that DNPC cells express genes defining basal, club, and hillock epithelial cells from benign prostate. We identified KLF5 as a regulator of genes defining this mixed basal, club, and hillock cell identity in DNPC models. KLF5-mediated upregulation of RARG uncovered a DNPC sensitivity to growth inhibition by retinoic acid receptor agonists, which down-regulated KLF5 and up-regulated AR. These findings offer CRPC classifications based on prostate epithelial cell identities and nominate KLF5 and RARG as therapeutic targets for CRPC displaying a mixed basal, club, and hillock identity.
Collapse
Affiliation(s)
- Samuel P. Pitzen
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN55455
- Graduate Program in Molecular, Cellular, and Developmental Biology and Genetics, University of Minnesota, Minneapolis, MN55455
| | - Amber N. Rudenick
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN55455
| | - Yinjie Qiu
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN55455
| | - Weijie Zhang
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN55455
| | - Sarah A. Munro
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN55455
| | - Braedan M. McCluskey
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN55455
| | - Colleen Forster
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN55455
| | - Hannah E. Bergom
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN55455
- Department of Medicine, University of Minnesota, Masonic Cancer Center, Minneapolis, MN55455
| | - Atef Ali
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN55455
- Department of Medicine, University of Minnesota, Masonic Cancer Center, Minneapolis, MN55455
| | - Ella Boytim
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN55455
- Department of Medicine, University of Minnesota, Masonic Cancer Center, Minneapolis, MN55455
| | - John T. Lafin
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Simon Linder
- Division on Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands1066 CX
| | - Mazlina Ismail
- Department of Oncology, University College London Cancer Institute, London, United KingdomWC1E 6BT
| | - Wout Devlies
- Department of Urology, University Hospitals Leuven, Leuven 3000, Belgium
- Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, Leuven3000, Belgium
| | | | - Frank Claessens
- Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, Leuven3000, Belgium
| | - Steven Joniau
- Department of Urology, University Hospitals Leuven, Leuven 3000, Belgium
- Department of Development and Regeneration, Katholieke Universiteit Leuven, Leuven3000, Belgium
| | - Gerhardt Attard
- Department of Oncology, University College London Cancer Institute, London, United KingdomWC1E 6BT
- University College London Hospitals, LondonWC1E 6DN, United Kingdom
| | - Wilbert Zwart
- Division on Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands1066 CX
| | - Peter S. Nelson
- Division of Hematology and Oncology, University of Washington, Fred Hutchinson Cancer Center, SeattleWA98109
- Human Biology Division, Fred Hutchinson Cancer Center, SeattleWA98109
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA98195
| | - Yuzhuo Wang
- Department of Urologic Sciences, Faculty of Medicine, Vancouver Prostate Centre, University of British Columbia, Vancouver, BCV6H 3Z6, Canada
- Department of Experimental Therapeutics, British Columbia Cancer Agency, Vancouver, BCV5Z 1L3, Canada
| | - Joshua M. Lang
- Department of Medicine, University of Wisconsin-Madison, Madison, WI53792
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI53792
| | - Himisha Beltran
- Department of Medical Oncology, Dana Farber Cancer Institute and Harvard Medical School, Boston, MA02115
| | - Douglas Strand
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Emmanuel S. Antonarakis
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN55455
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN55455
- Department of Medicine, University of Minnesota, Masonic Cancer Center, Minneapolis, MN55455
| | - Justin Hwang
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN55455
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN55455
- Department of Medicine, University of Minnesota, Masonic Cancer Center, Minneapolis, MN55455
| | - Paari Murugan
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN55455
| | - R. Stephanie Huang
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN55455
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN55455
| | - Scott M. Dehm
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN55455
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN55455
- Department of Urology, University of Minnesota, Minneapolis, MN55455
| |
Collapse
|
29
|
Graham LS, Su LJ, Nicklawsky A, Feng FX, Orlicky D, Petraccione J, Salzmann-Sullivan M, Nordeen SK, Flaig TW. Cell Models of Castration Resistant and High Dose Testosterone-Resistant Prostate Cancer Recapitulate the Heterogeneity of Response Observed in Clinical Practice. Cancers (Basel) 2025; 17:593. [PMID: 40002188 PMCID: PMC11852443 DOI: 10.3390/cancers17040593] [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: 12/11/2024] [Revised: 01/27/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
The use of supraphysiologic testosterone, particularly when alternated with an anti-androgen agent in men with metastatic castration-resistant prostate cancer (CRPC), has demonstrated promising results in clinical trials. As the use of this therapy in clinical practice is more widely adopted, there will be a growing need to understand the mechanisms of resistance. To that end, we independently derived three separate cell models of testosterone-sensitive CRPC. From each CRPC line, high dose testosterone-resistance (HTR) lines were selected. We demonstrated the differential response of the three CRPC lines to a high dose of testosterone in vitro and in vivo. We subsequently demonstrated the resistance of the HTR lines to testosterone and varying responses to testosterone withdrawal in vivo. The heterogeneity in responses to hormonal manipulation is correlated with varying levels of androgen receptor expression within the population. Overall, we show that we have developed three models of HTR that can be used to study the mechanisms of high dose testosterone resistance and identify potential therapeutic targets.
Collapse
Affiliation(s)
- Laura S. Graham
- Division of Medical Oncology, Department of Internal Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA; (L.-J.S.); (J.P.); (M.S.-S.); (T.W.F.)
| | - Lih-Jen Su
- Division of Medical Oncology, Department of Internal Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA; (L.-J.S.); (J.P.); (M.S.-S.); (T.W.F.)
| | - Andrew Nicklawsky
- Biostatistics and Bioinformatics, University of Colorado Cancer Center, Aurora, CO 80045, USA;
| | - Frances Xiuyan Feng
- Department of Pathology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA; (F.X.F.); (D.O.); (S.K.N.)
| | - David Orlicky
- Department of Pathology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA; (F.X.F.); (D.O.); (S.K.N.)
| | - Joseph Petraccione
- Division of Medical Oncology, Department of Internal Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA; (L.-J.S.); (J.P.); (M.S.-S.); (T.W.F.)
| | - Maren Salzmann-Sullivan
- Division of Medical Oncology, Department of Internal Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA; (L.-J.S.); (J.P.); (M.S.-S.); (T.W.F.)
| | - Steven K. Nordeen
- Department of Pathology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA; (F.X.F.); (D.O.); (S.K.N.)
| | - Thomas W. Flaig
- Division of Medical Oncology, Department of Internal Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA; (L.-J.S.); (J.P.); (M.S.-S.); (T.W.F.)
| |
Collapse
|
30
|
Haffner MC, Morris MJ, Ding CKC, Sayar E, Mehra R, Robinson B, True LD, Gleave M, Lotan TL, Aggarwal R, Huang J, Loda M, Nelson PS, Rubin MA, Beltran H. Framework for the Pathology Workup of Metastatic Castration-Resistant Prostate Cancer Biopsies. Clin Cancer Res 2025; 31:466-478. [PMID: 39589343 PMCID: PMC11790385 DOI: 10.1158/1078-0432.ccr-24-2061] [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: 07/01/2024] [Revised: 09/18/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Lineage plasticity and histologic transformation from prostate adenocarcinoma to neuroendocrine (NE) prostate cancer (NEPC) occur in up to 15% to 20% of patients with castration-resistant prostate cancer (CRPC) as a mechanism of treatment resistance and are associated with aggressive disease and poor prognosis. NEPC tumors typically display small cell carcinoma morphology with loss of androgen receptor (AR) expression and gain of NE lineage markers. However, there is a spectrum of phenotypes that are observed during the lineage plasticity process, and the clinical significance of mixed histologies or those that co-express AR and NE markers or lack all markers is not well defined. Translational research studies investigating NEPC have used variable definitions, making clinical trial design challenging. In this manuscript, we discuss the diagnostic workup of metastatic biopsies to help guide the reproducible classification of phenotypic CRPC subtypes. We recommend classifying CRPC tumors based on histomorphology (adenocarcinoma, small cell carcinoma, poorly differentiated carcinoma, other morphologic variant, or mixed morphology) and IHC markers with a priority for AR, NK3 homeobox 1, insulinoma-associated protein 1, synaptophysin, and cell proliferation based on Ki-67 positivity, with additional markers to be considered based on the clinical context. Ultimately, a unified workup of metastatic CRPC biopsies can improve clinical trial design and eventually practice.
Collapse
Affiliation(s)
- Michael C. Haffner
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Michael J. Morris
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chien-Kuang C. Ding
- Department of Anatomic Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Erolcan Sayar
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Rohit Mehra
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, Ann Arbor, MI, USA
- Rogel Cancer Center, Michigan Medicine, Ann Arbor, MI, USA
| | - Brian Robinson
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Lawrence D. True
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Martin Gleave
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Tamara L. Lotan
- Departments of Pathology, Urology, Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Rahul Aggarwal
- Division of Hematology/Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Jiaoti Huang
- Department of Pathology and Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Peter S. Nelson
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Mark A. Rubin
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Bern Center for Precision Medicine, University of Bern and Inselspital, Bern, Switzerland
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
31
|
Miyahira AK, Sharifi M, Chesner LN, El-Kenawi A, Haas R, Sena LA, Tewari AK, Pienta KJ, Soule HR. Personalized Medicine: Leave no Patient Behind; Report From the 2024 Coffey-Holden Prostate Cancer Academy Meeting. Prostate 2025; 85:211-226. [PMID: 39604057 DOI: 10.1002/pros.24826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024]
Abstract
INTRODUCTION The 11th Annual 2024 Coffey - Holden Prostate Cancer Academy (CHPCA) Meeting, was themed "Personalized Medicine: Leave No Patient Behind," and was held from June 20 to 23, 2024 at the University of California, Los Angeles, Luskin Conference Center, in Los Angeles, CA. METHODS The CHPCA Meeting is an academy-styled annual conference organized by the Prostate Cancer Foundation, to focus discussion on the most critical emerging research that have the greatest potential to advance knowledge of prostate cancer biology and treatment. The 2024 CHPCA Meeting was attended by 75 academic investigators and included 37 talks across 8 sessions. RESULTS The meeting sessions focused on: novel human, mouse and systems biology research models, novel immunotherapies for prostate cancer, efforts to overcome treatment resistance, the role of metabolism and diet in prostate cancer biology and as a therapeutic target, mechanisms that drive differentiation into neuroendocrine cancer subtypes, the evolving prostate cancer epigenome in disease progression and treatment resistance, and machine learning and advanced computational approaches for precision oncology. DISCUSSION This article summarizes the presentations and discussions from the 2024 CHPCA Meeting. We hope that sharing this knowledge will inspire and accelerate research into new discoveries and solutions for prostate cancer.
Collapse
Affiliation(s)
- Andrea K Miyahira
- Department of Science, Prostate Cancer Foundation, Santa Monica, California, USA
| | - Marina Sharifi
- Department of Medicine and Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Lisa N Chesner
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California, USA
| | - Asmaa El-Kenawi
- Department of Urology, Indiana University School of Medicine, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, Indiana, USA
| | - Roni Haas
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, California, USA
| | - Laura A Sena
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Alok K Tewari
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Kenneth J Pienta
- The James Buchanan Brady Urological Institute, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Howard R Soule
- Department of Science, Prostate Cancer Foundation, Santa Monica, California, USA
| |
Collapse
|
32
|
Poluben L, Nouri M, Liang J, Chen S, Varkaris A, Ersoy-Fazlioglu B, Voznesensky O, Lee II, Qiu X, Cato L, Seo JH, Freedman ML, Sowalsky AG, Lack NA, Corey E, Nelson PS, Brown M, Long HW, Russo JW, Balk SP. Increased nuclear factor I-mediated chromatin access drives transition to androgen receptor splice variant dependence in prostate cancer. Cell Rep 2025; 44:115089. [PMID: 39709604 PMCID: PMC11921039 DOI: 10.1016/j.celrep.2024.115089] [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: 04/04/2024] [Revised: 09/26/2024] [Accepted: 11/27/2024] [Indexed: 12/24/2024] Open
Abstract
Androgen receptor (AR) splice variants, of which ARv7 is the most common, are increased in castration-resistant prostate cancer, but the extent to which they drive AR activity is unclear. We generated a subline of VCaP cells (VCaP16) that is resistant to the AR inhibitor enzalutamide (ENZ). AR activity in VCaP16 is driven by ARv7, independently of full-length AR (ARfl), and its cistrome and transcriptome mirror those of ARfl in VCaP cells. ARv7 expression increases rapidly in response to ENZ, but there is a delay in gaining chromatin binding and transcriptional activity, which is associated with increased chromatin accessibility. AR and nuclear factor I (NFI) motifs are most enriched at more accessible sites, and NFIB/X knockdown greatly diminishes ARv7 function. These findings indicate that ARv7 can drive the AR program but that its activity is dependent on adaptations that increase chromatin accessibility to enhance its intrinsically weak chromatin binding.
Collapse
Affiliation(s)
- Larysa Poluben
- Department of Medicine and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Mannan Nouri
- Department of Medicine and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Jiaqian Liang
- Department of Medicine and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Shaoyong Chen
- Department of Medicine and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Andreas Varkaris
- Department of Medicine and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Betul Ersoy-Fazlioglu
- Department of Medicine and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Olga Voznesensky
- Department of Medicine and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Irene I Lee
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Xintao Qiu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Laura Cato
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Ji-Heui Seo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Matthew L Freedman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA; Eli and Edythe L. Broad Institute, Cambridge, MA, USA
| | - Adam G Sowalsky
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, Bethesda, MD, USA
| | - Nathan A Lack
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada; Department of Medical Pharmacology, School of Medicine, Koç University, Istanbul 34450, Turkey; Koç University Research Centre for Translational Medicine (KUTTAM), Koç University, Istanbul 34450, Turkey
| | - Eva Corey
- Department of Urology, University of Washington School of Medicine, Seattle, WA, USA
| | - Peter S Nelson
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Myles Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Henry W Long
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Joshua W Russo
- Department of Medicine and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| | - Steven P Balk
- Department of Medicine and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
33
|
MacDonald WJ, Purcell C, Pinho-Schwermann M, Stubbs NM, Srinivasan PR, El-Deiry WS. Heterogeneity in Cancer. Cancers (Basel) 2025; 17:441. [PMID: 39941808 PMCID: PMC11816170 DOI: 10.3390/cancers17030441] [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: 12/19/2024] [Revised: 01/21/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
Cancer heterogeneity is a major challenge in oncology, complicating diagnosis, prognostication, and treatment. The clinical heterogeneity of cancer, which leads to differential treatment outcomes between patients with histopathologically similar cancers, is attributable to molecular diversity manifesting through genetic, epigenetic, transcriptomic, microenvironmental, and host biology differences. Heterogeneity is observed between patients, individual metastases, and within individual lesions. This review discusses clinical implications of heterogeneity, emphasizing need for personalized approaches to overcome challenges posed by cancer's diverse presentations. Understanding of emerging molecular diagnostic and analytical techniques can provide a view into the multidimensional complexity of cancer heterogeneity. With over 90% of cancer-related deaths associated with metastasis, we additionally explore the role heterogeneity plays in treatment resistance and recurrence of metastatic lesions. Molecular insights from next-generation sequencing, single-cell transcriptomics, liquid biopsy technology, and artificial intelligence will facilitate the development of combination therapy regimens that can potentially induce lasting and even curative treatment outcomes.
Collapse
Affiliation(s)
- William J. MacDonald
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (W.J.M.); (M.P.-S.); (N.M.S.)
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Connor Purcell
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (W.J.M.); (M.P.-S.); (N.M.S.)
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Maximilian Pinho-Schwermann
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (W.J.M.); (M.P.-S.); (N.M.S.)
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Nolan M. Stubbs
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (W.J.M.); (M.P.-S.); (N.M.S.)
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Praveen R. Srinivasan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (W.J.M.); (M.P.-S.); (N.M.S.)
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Wafik S. El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (W.J.M.); (M.P.-S.); (N.M.S.)
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- The Joint Program in Cancer Biology, Brown University and Brown University Health, Providence, RI 02903, USA
- Hematology-Oncology Division, Department of Medicine, Rhode Island Hospital, Brown University, Providence, RI 02903, USA
| |
Collapse
|
34
|
Oyende Y, Taus LJ, Fatatis A. IL-1β in Neoplastic Disease and the Role of Its Tumor-Derived Form in the Progression and Treatment of Metastatic Prostate Cancer. Cancers (Basel) 2025; 17:290. [PMID: 39858071 PMCID: PMC11763358 DOI: 10.3390/cancers17020290] [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: 12/20/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Since its discovery, IL-1β has taken center stage as a key mediator of a very broad spectrum of diseases revolving around immuno-mediated and inflammatory events. Predictably, the pleiotropic nature of this cytokine in human pathology has led to the development of targeted therapeutics with multiple treatment indications in the clinic. Following the accumulated findings of IL-1β's central modulatory role in the immune system and the implication of inflammatory pathways in cancer, the use of IL-1β antagonists was first proposed and then also pursued for oncology disorders. However, this approach has consistently relied on the perceived need of interfering with IL-1β synthesized and secreted by immune cells. Herein, we discuss the importance of IL-1β derived from cancer cells which impacts primary tumors, particularly metastatic lesions, separately from and in addition to its more recognized role in immune-mediated inflammatory events. To this end, we focus on the instrumental contribution of IL-1β in the establishment and progression of advanced prostate adenocarcinoma. Special emphasis is placed on the potential role that the standard-of-care treatment strategies for prostate cancer patients have in unleashing IL-1β expression and production at metastatic sites. We conclude by reviewing the therapeutics currently used for blocking IL-1β signaling and propose a rationale for their concomitant use with standard-of-care treatments to improve the clinical outcomes of advanced prostate cancer.
Collapse
Affiliation(s)
- Yetunde Oyende
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA; (Y.O.); (L.J.T.)
| | - Luke J. Taus
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA; (Y.O.); (L.J.T.)
| | - Alessandro Fatatis
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA; (Y.O.); (L.J.T.)
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
35
|
Masur J, Ratan A, Wierbilowicz K, Ayanambakkam A, Churchman ML, Graham LS, Grass GD, Gupta S, Kern SQ, King J, Myint Z, Rounbehler RJ, Salhia B, Singer EA, Zakharia Y, Paschal BM, Viscuse PV. Clinical and Genomic Features of Androgen Indifferent Prostate Cancer. Int J Mol Sci 2025; 26:679. [PMID: 39859392 PMCID: PMC11765751 DOI: 10.3390/ijms26020679] [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: 12/12/2024] [Revised: 01/07/2025] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
Androgen-indifferent prostate cancer (AIPC) is increasingly common and particularly lethal. Data describing these tumors are sparse, and AIPC remains a poorly understood malignancy. Utilizing the Oncology Research Information Exchange Network (ORIEN) database, we enriched for tumors with features of AIPC using previously described characteristics. Our AIPC cohort included three subgroups: aggressive variant prostate cancer (AVPC), neuroendocrine PC (NEPC), and double-negative PC (DNPC). Of 1496 total PC patients available for analysis, we identified 323 (22%) as MCRPC. Of those, 39 (12%) met AIPC criteria (17 AVPC, 13 NEPC, 9 DNPC) and 284 (88%) were non-AIPC. Forty-three percent of AIPC patients had de novo metastatic disease vs. 15% for non-AIPC (p = 0.003). Homologous recombination deficiency (HRD) and tumor mutational burden (TMB) did not differ between cohorts, but microsatellite instability scores (MSI) were significantly higher in AIPC (p = 0.019). Using Gene Set Enrichment Analysis (GSEA), we found that genes defining response to androgens and genes involved in oxidative phosphorylation were the most downregulated, whereas genes involved in epithelial-mesenchymal transition (EMT) and immune signaling were significantly upregulated in AIPC vs. non-AIPC. Our study demonstrates the potential for predefined criteria that aim to enrich for AIPC and suggests opportunities for therapeutic investigation.
Collapse
Affiliation(s)
- Jack Masur
- Division of Hematology/Oncology, University of Virginia, Charlottesville, VA 22903, USA;
| | - Aakrosh Ratan
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA 22903, USA;
| | - Krzysztof Wierbilowicz
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA; (K.W.); (B.M.P.)
| | - Adanma Ayanambakkam
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | | | - Laura S. Graham
- Department of Medicine, Division of Medical Oncology, University of Colorado Cancer Center Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - George Daniel Grass
- H. Lee Moffitt Cancer Center, Department of Radiation Oncology, Tampa, FL 33612, USA;
| | - Sumati Gupta
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84103, USA;
| | - Sean Q. Kern
- Murtha Cancer Center, Uniformed Services University, Bethesda, MD 20814, USA;
| | - Jennifer King
- Department of Medicine, Division of Hematology and Medical Oncology, Indiana University Health Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA;
| | - Zin Myint
- Division of Medical Oncology, Department of Internal Medicine, Markey Cancer Center, University of Kentucky, Lexington, KY 40546, USA;
| | | | - Bodour Salhia
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA;
| | - Eric A. Singer
- Division of Urologic Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA;
| | - Yousef Zakharia
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA;
| | - Bryce M. Paschal
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA; (K.W.); (B.M.P.)
| | - Paul V. Viscuse
- Division of Hematology/Oncology, University of Virginia, Charlottesville, VA 22903, USA;
| |
Collapse
|
36
|
Mishra A, Huang SB, Dubash T, Burr R, Edd JF, Wittner BS, Cunneely QE, Putaturo VR, Deshpande A, Antmen E, Gopinathan KA, Otani K, Miyazawa Y, Kwak JE, Guay SY, Kelly J, Walsh J, Nieman LT, Galler I, Chan P, Lawrence MS, Sullivan RJ, Bardia A, Micalizzi DS, Sequist LV, Lee RJ, Franses JW, Ting DT, Brunker PAR, Maheswaran S, Miyamoto DT, Haber DA, Toner M. Tumor cell-based liquid biopsy using high-throughput microfluidic enrichment of entire leukapheresis product. Nat Commun 2025; 16:32. [PMID: 39746954 PMCID: PMC11696112 DOI: 10.1038/s41467-024-55140-x] [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: 04/12/2024] [Accepted: 11/28/2024] [Indexed: 01/04/2025] Open
Abstract
Circulating Tumor Cells (CTCs) in blood encompass DNA, RNA, and protein biomarkers, but clinical utility is limited by their rarity. To enable tumor epitope-agnostic interrogation of large blood volumes, we developed a high-throughput microfluidic device, depleting hematopoietic cells through high-flow channels and force-amplifying magnetic lenses. Here, we apply this technology to analyze patient-derived leukapheresis products, interrogating a mean blood volume of 5.83 liters from seven patients with metastatic cancer. High CTC yields (mean 10,057 CTCs per patient; range 100 to 58,125) reveal considerable intra-patient heterogeneity. CTC size varies within patients, with 67% overlapping in diameter with WBCs. Paired single-cell DNA and RNA sequencing identifies subclonal patterns of aneuploidy and distinct signaling pathways within CTCs. In prostate cancers, a subpopulation of small aneuploid cells lacking epithelial markers is enriched for neuroendocrine signatures. Pooling of CNV-confirmed CTCs enables whole exome sequencing with high mutant allele fractions. High-throughput CTC enrichment thus enables cell-based liquid biopsy for comprehensive monitoring of cancer.
Collapse
Affiliation(s)
- Avanish Mishra
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Shih-Bo Huang
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Howard Hughes Medical Institute, Bethesda, MD, 20815, USA
| | - Taronish Dubash
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Risa Burr
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Jon F Edd
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Ben S Wittner
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Quinn E Cunneely
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Victor R Putaturo
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Akansha Deshpande
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Ezgi Antmen
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Kaustav A Gopinathan
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Keisuke Otani
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Yoshiyuki Miyazawa
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Ji Eun Kwak
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Sara Y Guay
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Justin Kelly
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - John Walsh
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Linda T Nieman
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Isabella Galler
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - PuiYee Chan
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Michael S Lawrence
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Ryan J Sullivan
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Aditya Bardia
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
- Hematology/Oncology, University of California, Los Angeles, USA
| | - Douglas S Micalizzi
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Lecia V Sequist
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Richard J Lee
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Joseph W Franses
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - David T Ting
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Patricia A R Brunker
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Shyamala Maheswaran
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - David T Miyamoto
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA.
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
| | - Daniel A Haber
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA.
- Howard Hughes Medical Institute, Bethesda, MD, 20815, USA.
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA.
| | - Mehmet Toner
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
- Shriners Children's Boston, Boston, MA, 02114, USA.
| |
Collapse
|
37
|
Li F, Dai P, Shi H, Zhang Y, He J, Gopalan A, Li D, Chen Y, Du Y, Xu G, Yang W, Liang C, Gao D. LKB1 inactivation promotes epigenetic remodeling-induced lineage plasticity and antiandrogen resistance in prostate cancer. Cell Res 2025; 35:59-71. [PMID: 39743630 PMCID: PMC11701123 DOI: 10.1038/s41422-024-01025-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: 04/11/2024] [Accepted: 08/22/2024] [Indexed: 01/04/2025] Open
Abstract
Epigenetic regulation profoundly influences the fate of cancer cells and their capacity to switch between lineages by modulating essential gene expression, thereby shaping tumor heterogeneity and therapy response. In castration-resistant prostate cancer (CRPC), the intricacies behind androgen receptor (AR)-independent lineage plasticity remain unclear, leading to a scarcity of effective clinical treatments. Utilizing single-cell RNA sequencing on both human and mouse prostate cancer samples, combined with whole-genome bisulfite sequencing and multiple genetically engineered mouse models, we investigated the molecular mechanism of AR-independent lineage plasticity and uncovered a potential therapeutic strategy. Single-cell transcriptomic profiling of human prostate cancers, both pre- and post-androgen deprivation therapy, revealed an association between liver kinase B1 (LKB1) pathway inactivation and AR independence. LKB1 inactivation led to AR-independent lineage plasticity and global DNA hypomethylation during prostate cancer progression. Importantly, the pharmacological inhibition of TET enzymes and supplementation with S-adenosyl methionine were found to effectively suppress AR-independent prostate cancer growth. These insights shed light on the mechanism driving AR-independent lineage plasticity and propose a potential therapeutic strategy by targeting DNA hypomethylation in AR-independent CRPC.
Collapse
MESH Headings
- Male
- Animals
- Humans
- Epigenesis, Genetic/drug effects
- Mice
- Androgen Antagonists/pharmacology
- Androgen Antagonists/therapeutic use
- Protein Serine-Threonine Kinases/metabolism
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/drug effects
- Receptors, Androgen/metabolism
- Receptors, Androgen/genetics
- AMP-Activated Protein Kinase Kinases
- DNA Methylation/drug effects
- Cell Line, Tumor
- Prostatic Neoplasms, Castration-Resistant/pathology
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/metabolism
- Prostatic Neoplasms/pathology
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/drug therapy
- Prostatic Neoplasms/metabolism
- Cell Lineage
- Gene Expression Regulation, Neoplastic/drug effects
- Cell Plasticity/drug effects
- AMP-Activated Protein Kinases
Collapse
Affiliation(s)
- Fei Li
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Pengfei Dai
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Huili Shi
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yajuan Zhang
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juan He
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Anuradha Gopalan
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dan Li
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yu Chen
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yarui Du
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Guoliang Xu
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Weiwei Yang
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
| | - Chao Liang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Dong Gao
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China.
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China.
| |
Collapse
|
38
|
Nouruzi S, Zoubeidi A. LKB1 inactivation unleashes prostate cancer lineage plasticity. Cell Res 2025; 35:5-6. [PMID: 39743629 PMCID: PMC11701086 DOI: 10.1038/s41422-024-01030-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Affiliation(s)
- Shaghayegh Nouruzi
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
- Vancouver Prostate Centre, Vancouver, BC, V6H 3Z6, Canada
| | - Amina Zoubeidi
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada.
- Vancouver Prostate Centre, Vancouver, BC, V6H 3Z6, Canada.
| |
Collapse
|
39
|
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
|
40
|
Williams EC, Shibata M. Prostate Luminal Cell Plasticity and Cancer. Cancer Lett 2024:217430. [PMID: 39730086 DOI: 10.1016/j.canlet.2024.217430] [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: 07/29/2024] [Revised: 09/24/2024] [Accepted: 12/24/2024] [Indexed: 12/29/2024]
Abstract
Cellular plasticity in prostate cancer promotes treatment resistance. Several independent studies have used mouse models, single-cell RNA sequencing, and genetic lineage tracing approaches to characterize cellular differentiation and plasticity during prostate organogenesis, homeostasis and androgen-mediated tissue regeneration. We review these findings and recent work using immune-competent genetically-engineered mouse models to characterize cellular plasticity and clonal dynamic changes during prostate cancer progression. Collectively these studies highlight the influence of the tumor microenvironment and the function of epigenetic regulators in promoting cellular plasticity. How the epigenetic alternations that promote cell plasticity affect tumor immunogenicity remains an active area of research with implications for disease treatment.
Collapse
Affiliation(s)
- Emily C Williams
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA; The George Washington University Cancer Center, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Maho Shibata
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA; The George Washington University Cancer Center, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA.
| |
Collapse
|
41
|
Senatorov IS, Bowman J, Jansson KH, Alilin AN, Capaldo BJ, Lake R, Riba M, Abbey YC, Mcknight C, Zhang X, Raj S, Beshiri ML, Shinn P, Nguyen H, Thomas CJ, Corey E, Kelly K. Castrate-resistant prostate cancer response to taxane is determined by an HNF1-dependent apoptosis resistance circuit. Cell Rep Med 2024; 5:101868. [PMID: 39657662 PMCID: PMC11722106 DOI: 10.1016/j.xcrm.2024.101868] [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: 02/01/2024] [Revised: 09/09/2024] [Accepted: 11/14/2024] [Indexed: 12/12/2024]
Abstract
Metastatic castrate-resistant prostate cancer (mCRPC) is a genetically and phenotypically heterogeneous cancer where advancements are needed in biomarker discovery and targeted therapy. A critical and often effective component of treatment includes taxanes. We perform a high-throughput screen across a cohort of 30 diverse patient-derived castrate-resistant prostate cancer (CRPC) organoids to a library of 78 drugs. Combining quantitative response measures with transcriptomic analyses demonstrates that HNF1 homeobox A (HNF1A) drives a transcriptional program of taxane resistance, commonly dependent upon cellular inhibitor of apoptosis protein 2 (cIAP2). Monotherapy with cIAP2 inhibitor LCL161 is sufficient to treat HNF1A+ models of mCRPC previously resistant to docetaxel. These data may be useful in future clinical trial designs.
Collapse
Affiliation(s)
- Ilya S Senatorov
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Joel Bowman
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Keith H Jansson
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Aian Neil Alilin
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Brian J Capaldo
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Ross Lake
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Morgan Riba
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Yasmine C Abbey
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Crystal Mcknight
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Xiaohu Zhang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Sonam Raj
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Michael L Beshiri
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Paul Shinn
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Holly Nguyen
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Craig J Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA; Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, NIH, Bethesda, MD, USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Kathleen Kelly
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA.
| |
Collapse
|
42
|
Archer M, Lin KM, Kolanukuduru KP, Zhang J, Ben-David R, Kotula L, Kyprianou N. Impact of cell plasticity on prostate tumor heterogeneity and therapeutic response. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2024; 12:331-351. [PMID: 39839748 PMCID: PMC11744350 DOI: 10.62347/yfrp8901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 12/11/2024] [Indexed: 01/23/2025]
Abstract
Epithelial-mesenchymal transition (EMT) is a dynamic process of lineage plasticity in which epithelial cancer cells acquire mesenchymal traits, enabling them to metastasize to distant organs. This review explores the current understanding of how lineage plasticity and phenotypic reprogramming drive prostate cancer progression to lethal stages, contribute to therapeutic resistance, and highlight strategies to overcome the EMT phenotype within the prostate tumor microenvironment (TME). Emerging evidence reveals that prostate tumor cells can undergo lineage switching, adopting alternative growth pathways in response to anti-androgen therapies and taxane-based chemotherapy. These adaptive mechanisms support tumor survival and growth, underscoring the need for deeper insights into the processes driving prostate cancer differentiation, including neuroendocrine differentiation and lineage plasticity. A comprehensive understanding of these mechanisms will pave the way for innovative therapeutic strategies. Effectively targeting prostate cancer cells with heightened plasticity and therapeutic vulnerability holds promise for overcoming treatment resistance and preventing tumor recurrence. Such advancements are critical for developing effective approaches to prostate cancer treatment and improving patient survival outcomes.
Collapse
Affiliation(s)
- Maddison Archer
- Department of Urology, Icahn School of Medicine at Mount SinaiNew York, NY, USA
| | - Kevin M Lin
- Department of Urology, SUNY Upstate Medical UniversitySyracuse, NY, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical UniversitySyracuse, NY, USA
- Upstate Cancer Center, SUNY Upstate Medical UniversitySyracuse, NY, USA
| | | | - Joy Zhang
- Department of Urology, SUNY Upstate Medical UniversitySyracuse, NY, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical UniversitySyracuse, NY, USA
| | - Reuben Ben-David
- Department of Urology, Icahn School of Medicine at Mount SinaiNew York, NY, USA
| | - Leszek Kotula
- Department of Urology, SUNY Upstate Medical UniversitySyracuse, NY, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical UniversitySyracuse, NY, USA
- Upstate Cancer Center, SUNY Upstate Medical UniversitySyracuse, NY, USA
| | - Natasha Kyprianou
- Department of Urology, Icahn School of Medicine at Mount SinaiNew York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount SinaiNew York, NY, USA
- Department of Pathology and Molecular & Cell Based Medicine, Icahn School of Medicine at Mount SinaiNew York, NY, USA
| |
Collapse
|
43
|
Huang RR, Zuo C, Mona CE, Holzgreve A, Morrissey C, Nelson PS, Brady L, True L, Sisk A, Czernin J, Calais J, Ye H. FAP and PSMA Expression by Immunohistochemistry and PET Imaging in Castration-Resistant Prostate Cancer: A Translational Pilot Study. J Nucl Med 2024; 65:1952-1958. [PMID: 39477498 PMCID: PMC11619584 DOI: 10.2967/jnumed.124.268037] [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: 05/08/2024] [Accepted: 09/25/2024] [Indexed: 12/08/2024] Open
Abstract
Prostate-specific membrane antigen (PSMA) is a theranostic target for metastatic prostate cancer (PCa). However, castration-resistant PCa (CRPC) may lose PSMA expression after systemic therapy. Fibroblast activation protein (FAP), expressed by carcinoma-associated fibroblasts in various cancer types, including PCa, has the potential to be an alternative target. In this study, we evaluated FAP expression in CRPC to assess its potential, using PSMA as a comparison. Methods: FAP expression was assessed using immunohistochemistry in 116 CRPC tumors: 78 adenocarcinomas, 11 small cell carcinomas, and 27 anaplastic carcinomas. Correlation analysis between manual scoring and automated scoring was performed on 54 whole-slide sections of metastatic CRPC. Paired FAP and PSMA stains were assessed in tissue microarray cores of CRPC (n = 62), consisting of locally advanced CRPC (n = 9) and metastatic CRPC (n = 53). FAP and PSMA positivity was defined by an immunohistochemistry score of at least 10. To explore the correlation of PSMA and FAP inhibitor (FAPi) PET imaging and immunohistochemistry, a preliminary analysis of 4 patients included in a [68Ga]-FAPi-46 imaging trial (NCT04457232) was conducted. Results: Manual and automated scoring of FAP yielded results with strong correlations. Overall, FAP expression in CRPC was notably lower than PSMA expression (median immunoscores, 14 vs. 72; P < 0.001). Different histologic subtypes of CRPC demonstrated distinct levels of PSMA expression, whereas their FAP expression levels were comparable. Among the 19 PSMA-negative tumors, 11 (58%) exhibited FAP positivity. FAP expression levels in lymph node metastases were significantly lower than those in nonnodal metastases (P = 0.021). Liver metastases showed significant enrichment of tumors with strong FAP expression compared with nonliver lesions (P = 0.016). In the 4 clinical trial patients, the biopsied metastatic lesions showed lower uptake on FAPi PET than on PSMA PET (median SUVmax, 9.6 vs. 14.5), consistent with FAP expression that was lower than PSMA expression in the corresponding tumor biopsy samples (median immunoscores, 30 vs. 160). Conclusion: Because of the low FAP expression levels in CRPC, the utility of FAPi PET imaging may be limited. Although FAPi PET imaging may be further tested in PSMA-negative CRPC, such as small cell carcinoma, other molecular imaging modalities should be evaluated as alternative choices.
Collapse
Affiliation(s)
- Rong Rong Huang
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Chunlai Zuo
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California
- Department of Pathology, Rocky Mountain Regional VA Medical Center, Aurora, Colorado
| | - Christine E Mona
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Adrien Holzgreve
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, Washington
| | - Peter S Nelson
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Lauren Brady
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Lawrence True
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington; and
| | - Anthony Sisk
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Johannes Czernin
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Jeremie Calais
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, California;
| | - Huihui Ye
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California;
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
44
|
Nakanishi S, Suda T, Tanaka K, Yonamine T, Numahata K, Sugawa A, Oshiro T, Oshiro Y, Saito S, Inokuchi J. MUC1 expression is associated with ST3GAL2 and negatively correlated with the androgen receptor in castration-resistant prostate cancer. Glycoconj J 2024; 41:381-394. [PMID: 39718721 PMCID: PMC11735536 DOI: 10.1007/s10719-024-10173-8] [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: 08/09/2024] [Revised: 11/29/2024] [Accepted: 12/05/2024] [Indexed: 12/25/2024]
Abstract
Stage-specific embryonic antigen-4 (SSEA-4) is a developmentally regulated antigen, while expression level of SSEA-4 and / or its synthase ST3GAL2 is associated with prognosis in various malignancies. We have reported a prominent increase of SSEA-4 in castration-resistant prostate cancer (CRPC) and its negative correlation with the androgen receptor (AR). Meanwhile, loss of AR has increased to approximately 30% with the growing use of androgen receptor signaling inhibitor for metastatic CRPC (mCRPC). However, monitoring the progression status of AR-negative prostate cancer is a challenge because it does not produce prostate-specific antigen. Based on the negative relationship of expression between AR and SSEA-4, we hypothesized that a soluble molecule synchronized with SSEA-4 in expression could be a serum marker candidate for AR-negative prostate cancer. Thus, we investigated the molecular background of SSEA-4 expression by ST3GAL2-knockout in DU145 cells. Here we show that MUC1 is identified as a molecule associated with ST3GAL2 and expressed in AR-negative prostate cancer. A negative correlation of expression between AR and MUC1 was observed in prostate cancer cell lines and CRPC tissues. The average rate of MUC1 expression was nearly 60% in AR-negative prostate cancer cells in CRPC tissues. Level of serum CA15-3 (MUC1) was the highest in mCRPC among various stages and its higher level was associated with faster progression of mCRPC. Our results demonstrate that MUC1 is identified as a ST3GAL2-associated molecule and expressed in AR-negative CRPC cells. Furthermore, level of serum CA15-3 may reflect the progression status of mCRPC.
Collapse
Affiliation(s)
- Shotaro Nakanishi
- Department of Urology, University of the Ryukyus Graduate School of Medicine, 207 Uehara, Nishihara, Nakagami-gun, Okinawa, 903-0215, Japan
| | - Tetsuji Suda
- Department of Urology, University of the Ryukyus Graduate School of Medicine, 207 Uehara, Nishihara, Nakagami-gun, Okinawa, 903-0215, Japan
| | - Kei Tanaka
- Department of Urology, University of the Ryukyus Graduate School of Medicine, 207 Uehara, Nishihara, Nakagami-gun, Okinawa, 903-0215, Japan
| | - Tomoko Yonamine
- Department of Urology, University of the Ryukyus Graduate School of Medicine, 207 Uehara, Nishihara, Nakagami-gun, Okinawa, 903-0215, Japan
| | - Kenji Numahata
- Department of Urology, Yamagata Prefectural Central Hospital, 1800 Aoyagi, Yamagata, 990-2292, Japan
| | - Ai Sugawa
- Department of Urology, University of the Ryukyus Graduate School of Medicine, 207 Uehara, Nishihara, Nakagami-gun, Okinawa, 903-0215, Japan
| | - Takuma Oshiro
- Department of Urology, Naha City Hospital, 2-31-1 Furujima, Naha, 902-8511, Japan
| | - Yoshinori Oshiro
- Department of Urology, Chubu Tokushukai Hospital, 801 Higa, Kitanakagusuku-son, Nakagami-gun, Okinawa, 901-2393, Japan
| | - Seiichi Saito
- Department of Urology, University of the Ryukyus Graduate School of Medicine, 207 Uehara, Nishihara, Nakagami-gun, Okinawa, 903-0215, Japan.
- Medical Corporation Yoshinkai, 123 Daido, Naha, Okinawa, 902-0066, Japan.
| | - Junichi Inokuchi
- Department of Urology, University of the Ryukyus Graduate School of Medicine, 207 Uehara, Nishihara, Nakagami-gun, Okinawa, 903-0215, Japan.
| |
Collapse
|
45
|
Chatterjee SS, Linares JF, Cid-Diaz T, Duran A, Khan MIK, Osrodek M, Brady NJ, Reina-Campos M, Marzio A, Venkadakrishnan VB, Bakht MK, Khani F, Mosquera JM, Robinson BD, Moyer J, Elemento O, Hsieh AC, Goodrich DW, Rickman DS, Beltran H, Moscat J, Diaz-Meco MT. Increased translation driven by non-canonical EZH2 creates a synthetic vulnerability in enzalutamide-resistant prostate cancer. Nat Commun 2024; 15:9755. [PMID: 39567499 PMCID: PMC11579030 DOI: 10.1038/s41467-024-53874-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 10/25/2024] [Indexed: 11/22/2024] Open
Abstract
Overcoming resistance to therapy is a major challenge in castration-resistant prostate cancer (CRPC). Lineage plasticity towards a neuroendocrine phenotype enables CRPC to adapt and survive targeted therapies. However, the molecular mechanisms of epigenetic reprogramming during this process are still poorly understood. Here we show that the protein kinase PKCλ/ι-mediated phosphorylation of enhancer of zeste homolog 2 (EZH2) regulates its proteasomal degradation and maintains EZH2 as part of the canonical polycomb repressive complex (PRC2). Loss of PKCλ/ι promotes a switch during enzalutamide treatment to a non-canonical EZH2 cistrome that triggers the transcriptional activation of the translational machinery to induce a transforming growth factor β (TGFβ) resistance program. The increased reliance on protein synthesis creates a synthetic vulnerability in PKCλ/ι-deficient CRPC.
Collapse
Affiliation(s)
- Shankha S Chatterjee
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Juan F Linares
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Tania Cid-Diaz
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Angeles Duran
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Mohd Imran K Khan
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Marta Osrodek
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Nicholas J Brady
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | | | - Antonio Marzio
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | | | - Martin K Bakht
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Francesca Khani
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Juan Miguel Mosquera
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Brian D Robinson
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jenna Moyer
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Olivier Elemento
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Andrew C Hsieh
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Genomic Sciences, University of Washington, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - David W Goodrich
- Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - David S Rickman
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jorge Moscat
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA.
| | - Maria T Diaz-Meco
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
46
|
Oka T, Hatano K, Tani M, Yoshimura A, Horibe Y, Liu Y, Sassi N, Okuda Y, Yamamoto A, Uemura T, Yamamichi G, Ishizuya YU, Yamamoto Y, Kato T, Kawashima A, Fujita K, Nonomura N. PSA Kinetics Affect Prognosis in Patients With Castration-resistant Prostate Cancer Treated With Enzalutamide. CANCER DIAGNOSIS & PROGNOSIS 2024; 4:706-714. [PMID: 39502601 PMCID: PMC11534038 DOI: 10.21873/cdp.10385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 11/08/2024]
Abstract
Background/Aim There is little evidence regarding the predictive value of prostate-specific antigen (PSA) kinetics in patients with castration-resistant prostate cancer treated with an androgen receptor signaling inhibitor. This study investigated the correlation between PSA kinetics and prognosis in patients with castration-resistant prostate cancer treated with enzalutamide. Patients and Methods We analyzed data from 103 patients who received enzalutamide as primary treatment for castration-resistant prostate cancer at our hospital, focusing on the associations between overall survival and PSA kinetics variables, such as maximal PSA response, PSA nadir, and time to PSA nadir. Results The median PSA level at the initiation of enzalutamide was 18.1 ng/ml (interquartile range=7.9-61.2 ng/ml). The median maximal PSA response rate was 88% (interquartile range 55-98), and the median PSA nadir was 1.84 (interquartile range (IQR)=0.38-14.7) ng/ml. The median time to PSA nadir was 19 (IQR=6-28.5) weeks. Maximal PSA response rate <90% [hazard ratio (HR)=2.28, 95% confidence interval (CI)=1.03-5.03, p=0.0413], PSA nadir >2 ng/ml (HR=2.30, 95%CI=1.05-5.07, p=0.0379), time to nadir <19 weeks (HR=2.48, 95%CI=1.15-5.35, p=0.0204) were all independently predictive of shortened overall survival even after adjusting for pre-treatment factors. Conclusion Maximal PSA response, PSA nadir, and time to PSA nadir correlated with survival in patients with castration-resistant prostate cancer receiving enzalutamide as a first-line therapy.
Collapse
Affiliation(s)
- Toshiki Oka
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Koji Hatano
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masaru Tani
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Akihiro Yoshimura
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yuki Horibe
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yutong Liu
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Nesrine Sassi
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yohei Okuda
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Akinaru Yamamoto
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Toshihiro Uemura
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Gaku Yamamichi
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Y U Ishizuya
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshiyuki Yamamoto
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Taigo Kato
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Atsunari Kawashima
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazutoshi Fujita
- Department of Urology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Norio Nonomura
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
47
|
Xiong X, Zhang S, Zhu W, Du J, Liao X, Hu S, Yang J, Zheng W, Qiu S, Xu H, Wei Q, Yang L. Androgen-ablative therapies inducing CXCL8 regulates mTORC1/SREBP2-dependent cholesterol biosynthesis to support progression of androgen receptor negative prostate cancer cells. Oncogene 2024; 43:3456-3468. [PMID: 39369166 DOI: 10.1038/s41388-024-03181-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/07/2024]
Abstract
Treatment with androgen-ablative therapies effectively inhibited androgen receptor (AR)-positive (AR+) prostate cancer (PCa) cell subtypes, but it resulted in an increase in AR-negative (AR-) PCa cell subtypes. The present study aimed to investigate the debated mechanisms responsible for the changing proportion of cell types, identifying CXCL8 as a synthetic essential effector of AR- PCa cells. AR- PCa cells were found to be susceptible to CXCL8 depletion or inhibition, which impaired their survival. Mechanistically, androgen-ablative therapies resulted in the suppression of AR signaling, leading to the upregulation of CXCL8 gene transcription. CXCL8, in turn, activated the mTORC1 pathway, which increased de novo cholesterol synthesis by activating sterol regulatory element-binding protein-2 (SREBP2). Together, these results suggested that the CXCL8-mTORC1-SREBP2 axis contributed to the exacerbation of tumorigenicity in AR- PCa cells under androgen-ablative therapies.
Collapse
Affiliation(s)
- Xingyu Xiong
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, SC, China
| | - Shiyu Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, SC, China
| | - Weizhen Zhu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, SC, China
| | - Jiajia Du
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, SC, China
| | - Xinyang Liao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, SC, China
| | - Siping Hu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, SC, China
| | - Jie Yang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, SC, China
| | - Weitao Zheng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, SC, China
| | - Shi Qiu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, SC, China
- Center of Biomedical Big Data, West China Hospital Sichuan University, Chengdu, SC, China
| | - Hang Xu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, SC, China.
| | - Qiang Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, SC, China.
| | - Lu Yang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, SC, China.
| |
Collapse
|
48
|
Miller CD, Likasitwatanakul P, Toye E, Hwang JH, Antonarakis ES. Current uses and resistance mechanisms of enzalutamide in prostate cancer treatment. Expert Rev Anticancer Ther 2024; 24:1085-1100. [PMID: 39275993 PMCID: PMC11499039 DOI: 10.1080/14737140.2024.2405103] [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: 08/10/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 09/16/2024]
Abstract
INTRODUCTION Prostate cancer continues to be a major cause of morbidity and mortality for men worldwide. Enzalutamide, a second-generation non-steroidal antiandrogen that blocks androgen receptor (AR) transcriptional activity, is a treatment for biochemically recurrent, metastatic, castration-sensitive, and castration-resistant tumors. Unfortunately, most patients ultimately develop resistance to enzalutamide, making long-term treatment with this agent challenging. AREAS COVERED We performed a literature search of PubMed without date restrictions to investigate the literature surrounding enzalutamide and discuss the current uses of enzalutamide, proposed mechanisms driving resistance, and summarize current efforts to mitigate this resistance. EXPERT OPINION Enzalutamide is an effective prostate cancer therapy that is currently used in biochemically recurrent and metastatic disease and for both castration-sensitive and castration-resistant tumors. Unfortunately, resistance to enzalutamide occurs in each of these scenarios. In the clinical setting, enzalutamide-resistant tumors are either AR-driven or AR-indifferent. AR-dependent resistance mechanisms include genomic or epigenomic events that result in enhanced AR signaling. Tumors that do not require AR signaling instead may depend on alternative oncogenic pathways. There are numerous strategies to mitigate enzalutamide resistance, including concurrent use of PARP inhibitors or immune therapies. Additional work is required to uncover novel approaches to treat patients in the enzalutamide-resistant setting.
Collapse
Affiliation(s)
- Carly D. Miller
- Masonic Cancer Center, University of Minnesota-Twin Cities, Minneapolis, MN
| | - Pornlada Likasitwatanakul
- Masonic Cancer Center, University of Minnesota-Twin Cities, Minneapolis, MN
- Department of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Eamon Toye
- Masonic Cancer Center, University of Minnesota-Twin Cities, Minneapolis, MN
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Justin H. Hwang
- Masonic Cancer Center, University of Minnesota-Twin Cities, Minneapolis, MN
| | | |
Collapse
|
49
|
Rhee JW, Adzavon YM, Sun Z. Stromal androgen signaling governs essential niches in supporting prostate development and tumorigenesis. Oncogene 2024; 43:3419-3425. [PMID: 39369165 PMCID: PMC11573710 DOI: 10.1038/s41388-024-03175-1] [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: 07/04/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 10/07/2024]
Abstract
Androgens and androgen receptor (AR) mediated signaling pathways are essential for prostate development, morphogenesis, growth, and regeneration. Early tissue recombination experiments showed that AR-deficient urogenital sinus mesenchyme combined with intact urogenital sinus epithelium failed to develop into a prostate, demonstrating a stem cell niche for mesenchymal AR in prostatic development. Androgen signaling remains critical for prostate maturation and growth during postnatal stages. Importantly, most primary prostate cancer (PCa) cells express the AR, and aberrant activation of AR directly promotes PCa development, growth, and progression. Therefore, androgen deprivation therapy (ADT) targeting the AR in PCa cells is the main treatment for advanced PCa. However, it eventually fails, leading to the development of castration-resistant PCa, an incurable disease. Given these clinical challenges, the oncogenic AR action needs to be reevaluated for developing new and effective therapies. Recently, an essential niche role of stromal AR was identified in regulating prostate development and tumorigenesis. Here, we summarize the latest discoveries of stromal AR niches and their interactions with prostatic epithelia. In combination with emerging clinical and experimental evidence, we specifically discuss several important and long-term unanswered questions regarding tumor niche roles of stromal AR and highlight future therapeutic strategies by co-targeting epithelial and stromal AR for treating advanced PCa.
Collapse
Affiliation(s)
- June-Wha Rhee
- Department of Medicine, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Yao Mawulikplimi Adzavon
- Department of Cell Biology, Department of Oncology, Montefiore Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Zijie Sun
- Department of Cell Biology, Department of Oncology, Montefiore Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
50
|
Zhang Y, Shi X, Shi M, Li J, Liu Q. Androgens and androgen receptor directly induce the thickening, folding, and vascularization of the seahorse abdominal dermal layer into a placenta-like structure responsible for male pregnancy via multiple signaling pathways. Int J Biol Macromol 2024; 279:135039. [PMID: 39197609 DOI: 10.1016/j.ijbiomac.2024.135039] [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: 07/28/2024] [Revised: 08/22/2024] [Accepted: 08/22/2024] [Indexed: 09/01/2024]
Abstract
Seahorses exhibit the unique characteristic of male pregnancy, which incubates numerous embryos in a brood pouch that plays an essential role in enhancing offspring survivability. The pot-belly seahorse (Hippocampus abdominalis) possesses the largest body size among seahorses and is a significant species in Chinese aquaculture. In this study, we revealed the cytological and morphological characteristics, as well as regulatory mechanisms, throughout the entire brood pouch development in H. abdominalis. The brood pouch originated from the abdominal dermis, extending towards the ventral midline. As the dermal layers thicken, the inner epithelium folds, the stroma loosens, and vascularization occurs, culminating in the formation of the brood pouch. Furthermore, through transcriptomic analysis of brood pouches at various developmental stages, 8 key genes (tgfb3, fgf2, wnt7a, pgf, mycn, tln2, jund, ccn4) closely related to the development of brood pouch were identified in the MAPK, Rap1, TGF-β, and Wnt signaling pathways. These genes were highly expressed in the pseudoplacenta and dermal layers at the newly formed stage as examined by in situ hybridization (ISH). The angiogenesis, densification of collagen fibers, and proliferation of fibroblasts and endothelial cells in seahorse brood pouch formation may be regulated by these genes and pathways. Additionally, the expression of the androgen receptor gene (ar) was significantly upregulated during the formation of the brood pouch, and ISH confirmed the expression of the ar gene in the dermis and pseudoplacenta of the brood pouch, highlighting its role in the developmental process. Androgen and flutamide (androgen receptor antagonist) treatments significantly accelerated the formation of the brood pouch and completely inhibited its occurrence respectively, concomitant to the upregulated expression of differentially expressed genes involved above signaling pathways. These findings demonstrated that formation of the brood pouch is determined by androgen and the androgen receptor activates the above signaling pathways in the brood pouch through the regulation of fgf2, tgfb3, pgf, and wnt7a. Interestingly, androgen even induced the formation of the brood pouch in females. We firstly elucidated the formation of the seahorse brood pouch, demonstrating that androgens and their receptors directly induce the thickening, folding, and vascularization of the abdominal dermal layer into a placenta-like structure through multiple signaling pathways. These findings provide foundational insights to further exploring the evolution of male pregnancy and adaptive convergence in viviparity across vertebrates.
Collapse
Affiliation(s)
- Yichao Zhang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture (CAS), Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China; School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266000, China; Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
| | - Xuehui Shi
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Meilun Shi
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266000, China
| | - Jun Li
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture (CAS), Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China; Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.
| | - Qinghua Liu
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture (CAS), Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China; Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.
| |
Collapse
|