1
|
Zhang L, Teng PC, Cavassani KA, Wang J, Grasso C, Watson J, Chen Z, Tu KH, Salumbides B, Rohena-Rivera K, Gevorkian L, Kim M, You S, Di Vizio D, Sandler HM, Daskivich T, Bhowmick NA, Freeman MR, Tseng HR, Chen JF, Posadas EM. Emerin Dysregulation Drives the Very-Small-Nuclear Phenotype and Lineage Plasticity That Associate with a Clinically Aggressive Subtype of Prostate Cancer. Clin Cancer Res 2025; 31:2034-2045. [PMID: 40063516 PMCID: PMC12079098 DOI: 10.1158/1078-0432.ccr-24-3660] [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: 11/04/2024] [Revised: 01/23/2025] [Accepted: 03/06/2025] [Indexed: 05/16/2025]
Abstract
PURPOSE Circulating tumor cells (CTC) with a very-small-nuclear phenotype (vsnCTC) in prostate cancer are characterized by nuclei smaller than 8.5 μm. Our previous studies established an association between vsnCTCs and visceral metastasis. Reduction of emerin (EMD), a nuclear envelope protein, contributes to prostate cancer metastasis and nuclear shape instability. In this study, we investigated the correlation between EMD expression and the vsnCTC phenotype and its clinical impact. EXPERIMENTAL DESIGN We analyzed CTCs from 93 patients with metastatic castration-resistant prostate cancer and categorized them as either vsnCTC+ or vsnCTC- and compared overall survival and progression-free survival. C4-2B, 22Rv1, and DU145 with EMD knockdown were developed and characterized by nuclear size and gene expression by gene set enrichment analysis. Abiraterone- and enzalutamide-resistant C4-2B cells were also characterized by nuclear size and EMD expression. RESULTS Patients who were vsnCTC+ had significantly worse overall survival and progression-free survival compared with patients who were vsnCTC-. EMD expression was markedly reduced in CTCs from patients who were vsnCTC+ compared with patients who were vsnCTC-, with a significant positive correlation between EMD expression and CTC nuclear size. EMD knockdown in prostate cancer cells resulted in smaller nuclei, enhanced invasion, and the upregulation of genes associated with lineage plasticity. Additionally, abiraterone- and enzalutamide-resistant C4-2B cells had smaller nuclei and lower EMD expression. vsnCTC+ cells also showed enhanced platinum sensitivity. CONCLUSIONS The presence of vsnCTCs represents a novel hallmark of an aggressive subtype of metastatic castration-resistant prostate cancer closely linked to EMD loss and lineage plasticity. These findings highlight the importance of EMD dysregulation in the vsn phenotype, disease progression, and therapeutic resistance in patients with prostate cancer.
Collapse
MESH Headings
- Humans
- Male
- Prostatic Neoplasms, Castration-Resistant/pathology
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/mortality
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Neoplastic Cells, Circulating/pathology
- Neoplastic Cells, Circulating/metabolism
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Phenotype
- Cell Line, Tumor
- Gene Expression Regulation, Neoplastic
- Cell Nucleus/pathology
- Cell Nucleus/genetics
- Aged
- Middle Aged
- Biomarkers, Tumor/genetics
- Prostatic Neoplasms/pathology
- Prostatic Neoplasms/genetics
- Nitriles
- Benzamides
- Prognosis
- Phenylthiohydantoin/analogs & derivatives
- Phenylthiohydantoin/pharmacology
- Cell Lineage/genetics
- Drug Resistance, Neoplasm/genetics
Collapse
Affiliation(s)
- Le Zhang
- Center for Uro-Oncology Research Excellence, Cedars-Sinai Cancer, Los Angeles, California
- Division of Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Pai-Chi Teng
- Division of Urology, Department of Surgery, Cardinal Tien Hospital, Fu Jen Catholic University, Taipei, Taiwan
| | - Karen A. Cavassani
- Center for Uro-Oncology Research Excellence, Cedars-Sinai Cancer, Los Angeles, California
- Division of Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Jasmine Wang
- Center for Uro-Oncology Research Excellence, Cedars-Sinai Cancer, Los Angeles, California
- Division of Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Catherine Grasso
- Center for Uro-Oncology Research Excellence, Cedars-Sinai Cancer, Los Angeles, California
- Division of Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Joshua Watson
- Center for Uro-Oncology Research Excellence, Cedars-Sinai Cancer, Los Angeles, California
- Division of Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Zijing Chen
- Center for Uro-Oncology Research Excellence, Cedars-Sinai Cancer, Los Angeles, California
- Division of Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Kai-Han Tu
- Center for Uro-Oncology Research Excellence, Cedars-Sinai Cancer, Los Angeles, California
- Division of Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Brenda Salumbides
- Center for Uro-Oncology Research Excellence, Cedars-Sinai Cancer, Los Angeles, California
- Division of Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Krizia Rohena-Rivera
- Center for Uro-Oncology Research Excellence, Cedars-Sinai Cancer, Los Angeles, California
- Division of Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Lilit Gevorkian
- Center for Uro-Oncology Research Excellence, Cedars-Sinai Cancer, Los Angeles, California
- Division of Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Minhyung Kim
- Center for Uro-Oncology Research Excellence, Cedars-Sinai Cancer, Los Angeles, California
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Sungyong You
- Center for Uro-Oncology Research Excellence, Cedars-Sinai Cancer, Los Angeles, California
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Dolores Di Vizio
- Center for Uro-Oncology Research Excellence, Cedars-Sinai Cancer, Los Angeles, California
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, California
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Howard M. Sandler
- Center for Uro-Oncology Research Excellence, Cedars-Sinai Cancer, Los Angeles, California
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Timothy Daskivich
- Center for Uro-Oncology Research Excellence, Cedars-Sinai Cancer, Los Angeles, California
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Neil A. Bhowmick
- Center for Uro-Oncology Research Excellence, Cedars-Sinai Cancer, Los Angeles, California
- Division of Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Michael R. Freeman
- Center for Uro-Oncology Research Excellence, Cedars-Sinai Cancer, Los Angeles, California
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, California
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Hsian-Rong Tseng
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, California
| | - Jie-Fu Chen
- Department of Pathology, Memorial-Sloan Kettering Cancer Center, New York, Ney York
| | - Edwin M. Posadas
- Center for Uro-Oncology Research Excellence, Cedars-Sinai Cancer, Los Angeles, California
- Division of Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, California
- Cancer Therapeutics Program, Cedars-Sinai Cancer, Los Angeles, California
| |
Collapse
|
2
|
Teng M, Guo J, Xu X, Ci X, Mo Y, Kohen Y, Ni Z, Chen S, Guo WY, Bakht M, Ku S, Sigouros M, Luo W, Macarios CM, Xia Z, Chen M, Ul Haq S, Yang W, Berlin A, van der Kwast T, Ellis L, Zoubeidi A, Zheng G, Ming J, Wang Y, Cui H, Lok BH, Raught B, Beltran H, Qin J, He HH. Circular RMST cooperates with lineage-driving transcription factors to govern neuroendocrine transdifferentiation. Cancer Cell 2025; 43:891-904.e10. [PMID: 40250444 DOI: 10.1016/j.ccell.2025.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/31/2025] [Accepted: 03/21/2025] [Indexed: 04/20/2025]
Abstract
Circular RNA (circRNA) is a class of noncoding RNA with regulatory potentials. Its role in the transdifferentiation of prostate and lung adenocarcinoma into neuroendocrine prostate cancer (NEPC) and small cell lung cancer (SCLC) remains unexplored. Here, we identified circRMST as an exceptionally abundant circRNA predominantly expressed in NEPC and SCLC, with strong conservation between humans and mice. Functional studies using shRNA, siRNA, CRISPR-Cas13, and Cas9 consistently demonstrate that circRMST is essential for tumor growth and the expression of ASCL1, a master regulator of neuroendocrine fate. Genetic knockout of Rmst in NEPC genetic engineered mouse models prevents neuroendocrine transdifferentiation, maintaining tumors in an adenocarcinoma state. Mechanistically, circRMST physically interacts with lineage transcription factors NKX2-1 and SOX2. Loss of circRMST induces NKX2-1 protein degradation through autophagy-lysosomal pathway and alters the genomic binding of SOX2, collectively leading to the loss of ASCL1 transcription.
Collapse
Affiliation(s)
- Mona Teng
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Jiacheng Guo
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Xin Xu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Xinpei Ci
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Yulin Mo
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Yakup Kohen
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Zuyao Ni
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Sujun Chen
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Wang Yuan Guo
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Martin Bakht
- Division of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Shengyu Ku
- Division of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Michael Sigouros
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Wenqin Luo
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | | | - Ziting Xia
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
| | - Moliang Chen
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Sami Ul Haq
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Wen Yang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Alejandro Berlin
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Theo van der Kwast
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Leigh Ellis
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Amina Zoubeidi
- Vancouver Prostate Centre, Vancouver, BC, Canada; Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Gang Zheng
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jie Ming
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuzhuo Wang
- Vancouver Prostate Centre, Vancouver, BC, Canada; Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Haissi Cui
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
| | - Benjamin H Lok
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Brian Raught
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Himisha Beltran
- Division of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA.
| | - Jun Qin
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China.
| | - Housheng Hansen He
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
3
|
Zhao Y, Ramesh N, Xu P, Sei E, Hu M, Bai S, Troncoso P, Aparicio AM, Logothetis CJ, Corn PG, Navin NE, Zurita AJ. Longitudinal Profiling of Circulating Tumor DNA Reveals the Evolutionary Dynamics of Metastatic Prostate Cancer during Serial Therapy. Cancer Res 2025; 85:1680-1695. [PMID: 39992716 PMCID: PMC12048292 DOI: 10.1158/0008-5472.can-24-1943] [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: 06/09/2024] [Revised: 10/21/2024] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
Treatment decisions in metastatic castration-resistant prostate cancer are mostly guided by clinical variables, but efforts to molecularly monitor the disease remain hampered by challenges in acquiring tumor tissue repeatedly. In this study, we simultaneously profiled the genome copy number and exome in longitudinal plasma circulating tumor DNA (ctDNA) acquired before, during, and upon progression to serial treatments with androgen signaling inhibitors and taxane chemotherapy from 60 patients with metastatic castration-resistant prostate cancer (2-10 samples per patient). The genomic data were used to delineate the clonal substructure and evolutionary dynamics of each patient, and an evolutionary dynamic index was developed to measure the longitudinal changes of the tumor subclones. Treatment with androgen signaling inhibitors resulted in greater subclonal selection and population structure changes than taxane treatment. The subclones that emerged in association with serial therapy resistance harbored recurrent aberrations in previously identified and new candidate genes, with particular enrichment in genes related to PI3K-AKT signaling. These findings indicate that the integration of detailed clinical and genomic data can provide a framework for future unbiased genomic applications for ctDNA in the clinic to enable precision medicine. Significance: Profiling of the genomic copy number changes and mutations in circulating tumor DNA collected longitudinally from prostate cancer patients receiving serial life-prolonging therapies elucidates evolutionary dynamics and identifies emerging resistant subclones.
Collapse
Affiliation(s)
- Yuehui Zhao
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Naveen Ramesh
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas
- Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ping Xu
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
- Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Emi Sei
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Min Hu
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shanshan Bai
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Patricia Troncoso
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ana M Aparicio
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Christopher J Logothetis
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Paul G Corn
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nicholas E Navin
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Amado J Zurita
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
4
|
Labaf M, Han W, Zhang S, Liu M, Patten ND, Li M, Patalano S, Macoska JA, Balk SP, Han D, Zarringhalam K, Cai C. Heterogeneous Responses to High-Dose Testosterone in Castration-Resistant Prostate Cancer Tumors with Mixed Rb-Proficient and Rb-Deficient Cells. Mol Cancer Ther 2025; 24:772-783. [PMID: 40116305 PMCID: PMC12046331 DOI: 10.1158/1535-7163.mct-24-0716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/14/2025] [Accepted: 02/12/2025] [Indexed: 03/23/2025]
Abstract
Androgen deprivation therapy remains a cornerstone in managing prostate cancer. However, its recurrence often leads to the more aggressive castration-resistant prostate cancer (CRPC). Although second-line androgen receptor signaling inhibition treatments such as enzalutamide and abiraterone are available, their effectiveness against CRPC is only transient. High-dose testosterone (Hi-T) has recently emerged as a promising treatment for CRPC, primarily through the suppression of E2F and MYC signaling. However, the roles of Rb family proteins in influencing this therapeutic response remain debated. In this study, we utilized a CRPC patient-derived xenograft model that includes both Rb pathway-proficient and -deficient cell populations based on the positive or negative expression of RB family genes. Single-cell RNA sequencing analysis revealed that Rb-proficient cells displayed a robust response to Hi-T, whereas Rb-deficient cells exhibited significant resistance. Notably, our analysis indicated increased enrichment of the hypoxia signature in the Rb-deficient cell population. Further studies in RB1-silenced CRPC cell lines showed that treatment with a hypoxia-inducible factor-1α inhibitor can restore the sensitivity of Rb-deficient cells to high-dose dihydrotestosterone treatment. In conclusion, our research provides new molecular insights into CRPC tumor cell responses to Hi-T and proposes a new strategy to resensitize Rb-deficient CRPC cells to Hi-T treatment.
Collapse
Affiliation(s)
- Maryam Labaf
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, Massachusetts
- Department of Mathematics, University of Massachusetts Boston, Boston, Massachusetts
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Wanting Han
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, Massachusetts
- Department of Biology, University of Massachusetts Boston, Boston, Massachusetts
- Human Biology Division, Fred Hutchinson Cancer Center, Washington
| | - Songqi Zhang
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, Massachusetts
- Department of Biology, University of Massachusetts Boston, Boston, Massachusetts
| | - Mingyu Liu
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, Massachusetts
- Department of Biology, University of Massachusetts Boston, Boston, Massachusetts
| | - Nolan D. Patten
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, Massachusetts
- Department of Biology, University of Massachusetts Boston, Boston, Massachusetts
| | - Muqing Li
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, Massachusetts
- Department of Biology, University of Massachusetts Boston, Boston, Massachusetts
- Division of Urology, Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Susan Patalano
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, Massachusetts
- Department of Biology, University of Massachusetts Boston, Boston, Massachusetts
| | - Jill A. Macoska
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, Massachusetts
- Department of Biology, University of Massachusetts Boston, Boston, Massachusetts
| | - Steven P. Balk
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Dong Han
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, Massachusetts
- Department of Biology, University of Massachusetts Boston, Boston, Massachusetts
| | - Kourosh Zarringhalam
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, Massachusetts
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Changmeng Cai
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, Massachusetts
- Department of Biology, University of Massachusetts Boston, Boston, Massachusetts
| |
Collapse
|
5
|
Tian C, Yang S, Zhang C, Zhu R, Chen C, Wang X, Zhang D, Sun Q, Xu H, Nie H, Zhang Y, Ji D, Tang J, Jin K, Sun Y. Dual Role of CRABP2 in Colorectal Cancer: Oncogenesis via Nuclear RB1 and Cytoplasmic AFG3L2/SLC25A39 Axis, While Limiting Liver Metastasis through Cytoplasmic AFG3L2/PINK1/Parkin-Mediated Mitophagy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500552. [PMID: 40305785 DOI: 10.1002/advs.202500552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/31/2025] [Indexed: 05/02/2025]
Abstract
Colorectal cancer (CRC) progression and metastasis involve numerous regulatory factors. Among these, cellular retinoic acid-binding protein 2 (CRABP2) has been implicated as both a tumor activator and suppressor. Here, it is aimed to clarify the role of CRABP2 in CRC growth and metastasis and explore the underlying molecular mechanisms mediating its cellular functions. Using both in vitro and in vivo models, including a colonocyte-specific CRABP2 conditional knockout mouse model (Crabp2ΔIEC) and a subcutaneous tumorigenesis assay in BALB/c nude mice, it is shown that nuclear CRABP2 enhances tumor growth by interacting with and downregulating the tumor suppressor RB1, whereas cytoplasmic CRABP2 suppresses CRC liver metastasis by interacting with AFG3L2 and promoting mitophagy. In addition, the AFG3L2-SLC25A39 axis is identified as a distinct mechanism by which cytoplasmic CRABP2 increases mitochondrial glutathione stability to promote cell proliferation independent of the nuclear RB1 pathway. Notably, analysis of tissue from CRC patients reveals that CRABP2 protein has distinct prognostic implications and functional roles in the progression and metastasis of CRC dependent on its subcellular localization. Ultimately, by elucidating the role of CRABP2 in CRC, it is aimed to provide new insight into disease pathogenesis and inform the development of therapeutic interventions.
Collapse
Affiliation(s)
- Chuanxin Tian
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Sheng Yang
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Chuan Zhang
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Renzhong Zhu
- Institute of Translational Medicine, Medical College, Yangzhou University, No.136 Jiangyang Road, Yangzhou, 210029, China
| | - Chen Chen
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Xiaowei Wang
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Dongsheng Zhang
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Qingyang Sun
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Hengjie Xu
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Hongxu Nie
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Yue Zhang
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Dongjian Ji
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Junwei Tang
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Kangpeng Jin
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Yueming Sun
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Saloni, Sachan M, Rahul, Verma RS, Patel GK. SOXs: Master architects of development and versatile emulators of oncogenesis. Biochim Biophys Acta Rev Cancer 2025; 1880:189295. [PMID: 40058508 DOI: 10.1016/j.bbcan.2025.189295] [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: 10/02/2024] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 03/22/2025]
Abstract
Transcription factors regulate a variety of events and maintain cellular homeostasis. Several transcription factors involved in embryonic development, has been shown to be closely associated with carcinogenesis when deregulated. Sry-like high mobility group box (SOX) proteins are potential transcription factors which are evolutionarily conserved. They regulate downstream genes to determine cell fate, via various signaling pathways and cellular processes essential for tissue and organ development. Dysregulation of SOXs has been reported to promote or suppress tumorigenesis by modulating cellular reprogramming, growth, proliferation, angiogenesis, metastasis, apoptosis, immune modulation, lineage plasticity, maintenance of the stem cell pool, therapy resistance and cancer relapse. This review provides a crucial understanding of the molecular mechanism by which SOXs play multifaceted roles in embryonic development and carcinogenesis. It also highlights their potential in advancing therapeutic strategies aimed at targeting SOXs and their downstream effectors in various malignancies.
Collapse
Affiliation(s)
- Saloni
- Cancer and Stem Cell Laboratory, Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211004, India
| | - Manisha Sachan
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211004, India
| | - Rahul
- Department of Surgical Gastroenterology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow 226014, India
| | - Rama Shanker Verma
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211004, India.
| | - Girijesh Kumar Patel
- Cancer and Stem Cell Laboratory, Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211004, India.
| |
Collapse
|
9
|
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
|
10
|
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
|
11
|
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
|
12
|
Lee J, Lee Y. The role of transcription factors in prostate cancer progression. Mol Cells 2025; 48:100193. [PMID: 39938868 PMCID: PMC11907451 DOI: 10.1016/j.mocell.2025.100193] [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: 11/14/2024] [Revised: 12/23/2024] [Accepted: 02/04/2025] [Indexed: 02/14/2025] Open
Abstract
Prostate cancer is one of the most common malignancies in men, with most cases initially responding to androgen deprivation therapy. However, a significant number of patients eventually develop castration-resistant prostate cancer, an aggressive form of the disease. Although androgen receptor (AR) pathway inhibitors target AR signaling, and have extended survival in patients with castration-resistant prostate cancer, prolonged treatment can lead to the emergence of neuroendocrine prostate cancer (NEPC), a lethal subtype characterized by the expression of neuroendocrine markers and reduced AR activity. The transition from adenocarcinoma to NEPC is driven by lineage plasticity, wherein cancer cells adopt a neuroendocrine phenotype to evade treatment. Consequently, NEPC patients face poor clinical outcomes and limited effective treatment options. To improve outcomes, it is crucial to understand the molecular mechanisms driving NEPC development. In this review, we highlight the role of transcription factors in this process and explore their potential as therapeutic targets.
Collapse
Affiliation(s)
- Jongeun Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Yoontae Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk 37673, Republic of Korea.
| |
Collapse
|
13
|
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
|
14
|
Lee J, Park J, Hur Y, Um D, Choi HS, Park J, Kim Y, Lee JS, Choi K, Kim E, Park YB, Choi JM, Kim TK, Lee Y. ETV5 reduces androgen receptor expression and induces neural stem-like properties during neuroendocrine prostate cancer development. Proc Natl Acad Sci U S A 2025; 122:e2420313122. [PMID: 40117308 PMCID: PMC11962414 DOI: 10.1073/pnas.2420313122] [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: 10/03/2024] [Accepted: 02/18/2025] [Indexed: 03/23/2025] Open
Abstract
Neuroendocrine prostate cancer (NEPC), an aggressive subtype induced by hormone therapy, lacks effective treatments. This study explored the role of E26 transformation-specific variant 5 (ETV5) in NEPC development. Analysis of multiple prostate cancer datasets revealed that NEPC is characterized by significantly elevated ETV5 expression compared to other subtypes. ETV5 expression increased progressively under hormone therapy through epigenetic modifications. ETV5 induced neural stem-like features in prostate cancer cells and facilitated their differentiation into NEPC under hormone treatment conditions, both in vitro and in vivo. Our molecular mechanistic study identified PBX3 and TLL1 as target genes of ETV5 that contribute to ETV5 overexpression-induced castration resistance and stemness. Notably, obeticholic acid, identified as an ETV5 inhibitor in this study, exhibited promising efficacy in suppressing NEPC development. This study highlights ETV5 as a key transcription factor that facilitates NEPC development and underscores its potential as a therapeutic target for this aggressive cancer subtype.
Collapse
Affiliation(s)
- Jongeun Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk37673, Republic of Korea
| | - Jiho Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk37673, Republic of Korea
| | - Yunjung Hur
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk37673, Republic of Korea
| | - Dahun Um
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk37673, Republic of Korea
| | - Hyung-Seok Choi
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk37673, Republic of Korea
| | - Joonyoung Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk37673, Republic of Korea
| | - Yewon Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk37673, Republic of Korea
| | - Jeon-Soo Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk37673, Republic of Korea
| | - Kyuha Choi
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk37673, Republic of Korea
| | - Eunjeong Kim
- Department of Biology, College of Natural Sciences, Kyungpook National University, Daegu41566, Republic of Korea
| | - Young Bin Park
- Calici Co., Ltd., Korea, Daejeon34134, Republic of Korea
| | - Jae-Mun Choi
- Calici Co., Ltd., Korea, Daejeon34134, Republic of Korea
- Department of Bio-Artificial Intelligence Convergence, Chungnam National University, Daejeon34134, Republic of Korea
- Department of Food and Biotechnology, Korea University, Sejong30019, Republic of Korea
| | - Tae-Kyung Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk37673, Republic of Korea
| | - Yoontae Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk37673, Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul03722, Republic of Korea
| |
Collapse
|
15
|
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
|
16
|
Wang Y, Xue H, Zhu X, Lin D, Chen Z, Dong X, Chen J, Shi M, Ni Y, Cao J, Wu R, Kang C, Pang X, Crea F, Lin YY, Collins CC, Gleave ME, Parolia A, Chinnaiyan A, Ong CJ, Wang Y. Deciphering the Transcription Factor Landscape in Prostate Cancer Progression: A Novel Approach to Understand NE Transdifferentiation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2404938. [PMID: 40091506 DOI: 10.1002/advs.202404938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 02/18/2025] [Indexed: 03/19/2025]
Abstract
Prostate cancer (PCa) stands as a leading cause of cancer-related mortality among men, with treatment-induced neuroendocrine prostate cancer (NEPC) posing a challenge as an ARPI-resistant subtype. The role of transcription factors (TFs) in PCa progression and NEPC transdifferentiation remains inadequately understood, underscoring a critical gap in current research. In this study, an internal Z score-based approach is developed to identify lineage-specific TF profiles in prostatic adenocarcinoma and NEPC for a nuanced understanding of TF expression dynamics. Distinct TF profiles for adenocarcinoma and NEPC are unveiled, identifying 126 shared TFs, 46 adenocarcinoma-TFs, and 56 NEPC-TFs, validated across multiple cohorts. Gene Ontology is employed to validate their biological and functional roles in PCa progression. Implications are revealed in cell development, differentiation, and lineage determination. Knockdown experiments suggest that lineage-TFs are functionally important in maintaining lineage-specific cell proliferation. Additionally, a longitudinal study on NE transdifferentiation highlights dynamic TF expression shifts, proposing a three-phases hypothesis for PCa progression mechanisms. This study introduces a groundbreaking approach for deciphering the TF landscape in PCa, providing a molecular basis for adenocarcinoma to NEPC progression, and paving the way for innovative treatment strategies with potential impact on patient outcomes.
Collapse
Affiliation(s)
- Yu Wang
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, V5Z 1M9, Canada
- Vancouver Prostate Centre, Vancouver, V6H 3Z6, Canada
- Department of Experimental Therapeutics, BC Cancer, Vancouver, V5Z 1L3, Canada
| | - Hui Xue
- Department of Experimental Therapeutics, BC Cancer, Vancouver, V5Z 1L3, Canada
| | - Xiaohui Zhu
- The First Affiliated Hospital of Jinan University, First Clinical Medical College, Jinan University, Guangzhou, 510632, P. R. China
| | - Dong Lin
- Vancouver Prostate Centre, Vancouver, V6H 3Z6, Canada
- Department of Experimental Therapeutics, BC Cancer, Vancouver, V5Z 1L3, Canada
| | - Zheng Chen
- The First Affiliated Hospital of Jinan University, First Clinical Medical College, Jinan University, Guangzhou, 510632, P. R. China
| | - Xin Dong
- Department of Experimental Therapeutics, BC Cancer, Vancouver, V5Z 1L3, Canada
| | - Junru Chen
- Department of Urology, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Mingchen Shi
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, V5Z 1M9, Canada
- Vancouver Prostate Centre, Vancouver, V6H 3Z6, Canada
- Department of Experimental Therapeutics, BC Cancer, Vancouver, V5Z 1L3, Canada
| | - Yuchao Ni
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, V5Z 1M9, Canada
- Vancouver Prostate Centre, Vancouver, V6H 3Z6, Canada
- Department of Experimental Therapeutics, BC Cancer, Vancouver, V5Z 1L3, Canada
- Department of Urology, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Jonathan Cao
- Department of Cell and Systems Biology, University of Toronto, Toronto, M5S 3G5, Canada
| | - Rebecca Wu
- Department of Experimental Therapeutics, BC Cancer, Vancouver, V5Z 1L3, Canada
| | - Connie Kang
- Department of Experimental Therapeutics, BC Cancer, Vancouver, V5Z 1L3, Canada
| | - Xinyao Pang
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, V5Z 1M9, Canada
- Vancouver Prostate Centre, Vancouver, V6H 3Z6, Canada
- Department of Experimental Therapeutics, BC Cancer, Vancouver, V5Z 1L3, Canada
| | - Francesco Crea
- Cancer Research Group, School of Life Health and Chemical Sciences, The Open University, Milton Keynes, MK7 6AA, UK
| | - Yen-Yi Lin
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, V5Z 1M9, Canada
- Vancouver Prostate Centre, Vancouver, V6H 3Z6, Canada
| | - Colin C Collins
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, V5Z 1M9, Canada
- Vancouver Prostate Centre, Vancouver, V6H 3Z6, Canada
| | - Martin E Gleave
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, V5Z 1M9, Canada
- Vancouver Prostate Centre, Vancouver, V6H 3Z6, Canada
| | - Abhijit Parolia
- Michigan Center for Translational Pathology, Department of Urology, University of Michigan Medical School, Rogel Cancer Center, University of Michigan Hospital, Ann Arbor, 48109, USA
| | - Arul Chinnaiyan
- Michigan Center for Translational Pathology, Department of Urology, University of Michigan Medical School, Rogel Cancer Center, University of Michigan Hospital, Ann Arbor, 48109, USA
| | - Christopher J Ong
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, V5Z 1M9, Canada
- Vancouver Prostate Centre, Vancouver, V6H 3Z6, Canada
| | - Yuzhuo Wang
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, V5Z 1M9, Canada
- Vancouver Prostate Centre, Vancouver, V6H 3Z6, Canada
- Department of Experimental Therapeutics, BC Cancer, Vancouver, V5Z 1L3, Canada
| |
Collapse
|
17
|
Foster E, Wardhana O, Zeng Z, Lu X. Comparative Analysis of 3D Culture Methodologies in Prostate Cancer Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.11.642589. [PMID: 40161751 PMCID: PMC11952452 DOI: 10.1101/2025.03.11.642589] [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: 04/02/2025]
Abstract
Three-dimensional (3D) cell culture models are increasingly utilized in cancer research to better replicate in vivo tumor microenvironments. This study examines the effects of different 3D scaffolding materials, including Matrigel, GelTrex, and the plant-based GrowDex, on prostate cancer cell lines, with a particular emphasis on neuroendocrine prostate cancer (NEPC). Four cell lines (LNCaP, LASCPC-01, PC-3, and KUCaP13) were cultured in these scaffolds to evaluate spheroid formation, cell viability, and gene expression. The results revealed that while all scaffolds supported cell viability, spheroid formation varied significantly: Matrigel promoted the most robust spheroids, especially for LASCPC-01, whereas GrowDex exhibited limitations for certain cell lines. Gene expression analysis indicated a consistent reduction in androgen receptor (AR) expression in LNCaP cells across all scaffolds, suggesting a potential shift towards a neuroendocrine phenotype. However, the expression of neuroendocrine markers varied depending on the scaffold and culture method, with the mini-domes method in Matrigel leading to decreased expression of both castration-resistant prostate cancer (CRPC) and NEPC markers. These findings highlight the scaffold-dependent variability in 3D culture outcomes and emphasize the need for standardized methodologies to ensure consistency and relevance in prostate cancer research.
Collapse
Affiliation(s)
- Ella Foster
- Department of Biological Sciences, Boler-Parseghian Center for Rare and Neglected Diseases, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Oliver Wardhana
- Department of Biological Sciences, Boler-Parseghian Center for Rare and Neglected Diseases, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Ziyu Zeng
- Department of Biological Sciences, Boler-Parseghian Center for Rare and Neglected Diseases, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Xin Lu
- Department of Biological Sciences, Boler-Parseghian Center for Rare and Neglected Diseases, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
- Integrated Biomedical Sciences Graduate Program, University of Notre Dame, Notre Dame, IN, 46556, USA
- Tumor Microenvironment and Metastasis Program, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| |
Collapse
|
18
|
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
|
19
|
Fernandez-Cuesta L, Alcala N, Mathian E, Derks J, Thirlwell C, Dayton T, Marinoni I, Perren A, Walter T, Foll M. Basic science and translational implications of current knowledge on neuroendocrine tumors. J Clin Invest 2025; 135:e186702. [PMID: 40026252 PMCID: PMC11870734 DOI: 10.1172/jci186702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025] Open
Abstract
Neuroendocrine tumors (NETs) are a diverse group of malignancies that can occur in various organs, with a notable prevalence in the lungs and gastrointestinal tract, which are the focus of this Review. Although NETs are rare in individual organs, their incidence has increased over recent decades, highlighting the urgent need for current classification systems to evolve by incorporating recent advances in the understanding of NET biology. Several omics studies have revealed molecular subtypes, which, when integrated into existing classification frameworks, may provide more clinically relevant insights for patients with NETs. This Review examines recent progress in elucidating the biology of NETs, with a particular emphasis on the tumor microenvironment and cells of origin. The existence of different cells of origin, which may contribute to distinct molecular groups, along with profiles of immune infiltration - despite being generally low - could explain the emergence of more aggressive cases and the potential for metastatic progression. Given the molecular heterogeneity of NETs and the diversity of their microenvironments and different cells of origin, there is an urgent need to develop morphomolecular classification systems. Such systems would make it possible to better characterize tumor progression, identify new therapeutic targets, and, ultimately, guide the development of personalized therapies.
Collapse
Affiliation(s)
- Lynnette Fernandez-Cuesta
- Computational Cancer Genomics Team, Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Nicolas Alcala
- Computational Cancer Genomics Team, Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Emilie Mathian
- Computational Cancer Genomics Team, Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Jules Derks
- Department of Pulmonary Medicine, Erasmus MC Cancer institute, University Medical Center, Rotterdam, Netherlands
- GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | | | - Talya Dayton
- European Molecular Biology Laboratory Barcelona, Tissue Biology and Disease Modeling, Barcelona, Spain
| | - Ilaria Marinoni
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Aurel Perren
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Thomas Walter
- Service d’Oncologie Médicale, Groupement Hospitalier Centre, Institut de Cancérologie des Hospices Civils de Lyon, Lyon, France
| | - Matthieu Foll
- Computational Cancer Genomics Team, Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| |
Collapse
|
20
|
Uchida K, Fujisaki A, Imai S, Otsuki Y, Yoneda T. Rapidly Progressing Prostate Cancer With Low Prostate-Specific Antigen and Gleason Score 5+5: A Case Report. Cureus 2025; 17:e80808. [PMID: 40255806 PMCID: PMC12006871 DOI: 10.7759/cureus.80808] [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] [Accepted: 03/18/2025] [Indexed: 04/22/2025] Open
Abstract
An 87-year-old man presented with worsening urinary dysfunction with urinary retention. The prostate-specific antigen was 1.23 ng/mL. Prostate biopsy confirmed adenocarcinoma with a Gleason score of 5+5; the final diagnosis was T4N1M0. Androgen deprivation therapy and immediate whole-pelvic radiotherapy (80 Gy) were administered. Nadir prostate-specific antigen of 0.40 ng/mL two months later increased to 35.92 ng/mL as the disease metastasized to the corpus cavernosum penis, causing malignant priapism. The best supportive care was provided, and the patient died 11 months later. High-grade prostate cancers with low prostate-specific antigen levels are rare but highly aggressive. Androgen deprivation therapy and radiotherapy alone may be insufficient in such cases, highlighting the need for more effective treatment strategies.
Collapse
Affiliation(s)
- Kosuke Uchida
- Department of Urology, Seirei Hamamatsu General Hospital, Hamamatsu, JPN
| | - Akira Fujisaki
- Department of Urology, Seirei Hamamatsu General Hospital, Hamamatsu, JPN
| | - Shin Imai
- Department of Urology, Seirei Hamamatsu General Hospital, Hamamatsu, JPN
| | - Yoshiro Otsuki
- Department of Pathology, Seirei Hamamatsu General Hospital, Hamamatsu, JPN
| | - Tatsuaki Yoneda
- Department of Urology, Seirei Hamamatsu General Hospital, Hamamatsu, JPN
| |
Collapse
|
21
|
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
|
22
|
Zhang X, Yang Y, Zou H, Yang Y, Zheng X, Corey E, Zoubeidi A, Mitsiades N, Yu AM, Li Y, Chen HW. Effective therapeutic targeting of tumor lineage plasticity in neuroendocrine prostate cancer by BRD4 inhibitors. Acta Pharm Sin B 2025; 15:1415-1429. [PMID: 40370549 PMCID: PMC12069891 DOI: 10.1016/j.apsb.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/13/2024] [Accepted: 11/18/2024] [Indexed: 05/16/2025] Open
Abstract
Tumor lineage plasticity (LP) is an emerging hallmark of cancer progression. Through pharmacologically probing the function of epigenetic regulators in prostate cancer cells and organoids, we identified bromodomain protein BRD4 as a crucial player. Integrated ChIP-seq and RNA-seq analysis of tumors revealed, for the first time, that BRD4 directly activates hundreds of genes in the LP programs which include neurogenesis, axonogenesis, EMT and stem cells and key drivers such as POU3F2 (BRN2), ASCL1/2, NeuroD1, SOX2/9, RUNX1/2 and DLL3. Interestingly, BRD4 genome occupancy is reprogrammed by anti-AR drugs from facilitating AR function in CRPC cells to activating the LP programs and is facilitated by pioneer factor FOXA1. Significantly, we demonstrated that BRD4 inhibitor AZD5153, currently at clinical development, possesses potent activities in complete blockade of tumor growth of both de novo neuroendocrine prostate cancer (NEPC) and treatment-induced NEPC PDXs and that suppression of tumor expression of LP programs through reduction of local chromatin accessibility is the primary mechanism of action (MOA) by AZD5153. Together, our study revealed that BRD4 plays a fundamental role in direct activation of tumor LP programs and that its inhibitor AZD5153 is highly promising in effective treatment of the lethal forms of the diseases.
Collapse
Affiliation(s)
- Xiong Zhang
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Yatian Yang
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Hongye Zou
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Yang Yang
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Xingling Zheng
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, DC 98915, USA
| | - Amina Zoubeidi
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC V5Z1M9, Canada
| | - Nicolas Mitsiades
- Department of Internal Medicine, Division of Hematology and Oncology, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Ai-Ming Yu
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Yuanpei Li
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Hong-Wu Chen
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
- Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
- VA Northern California Health Care System-Mather, Mather, CA 95655, USA
| |
Collapse
|
23
|
Huang F, Li K, Chen Z, Cui Z, Hankey W, Fang K, Yan J, Wang H, Jin VX, Dong Y, Wang Q. Integrative analysis identifies the atypical repressor E2F8 as a targetable transcriptional activator driving lethal prostate cancer. Oncogene 2025; 44:481-493. [PMID: 39613933 DOI: 10.1038/s41388-024-03239-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/12/2024] [Accepted: 11/22/2024] [Indexed: 12/01/2024]
Abstract
Acquired resistance to androgen receptor (AR)-targeted therapies underscores the need to identify alternative therapeutic targets for treating lethal prostate cancer. In this study, we evaluated the prognostic significance of 1635 human transcription factors (TFs) by analyzing castration-resistant prostate cancer (CRPC) datasets from the West and East Stand Up to Cancer (SU2C) cohorts. Through this screening approach, we identified E2F8, a putative transcriptional repressor, as a TF consistently associated with poorer patient outcomes in both cohorts. Notably, E2F8 is highly expressed and active in AR-negative CRPC compared to AR-positive CRPC. Integrative profiling of E2F8 cistromes and transcriptomes in AR-negative CRPC cells revealed that E2F8 directly and non-canonically activates target oncogenes involved in cancer-associated pathways. To target E2F8 in CRPC, we employed the CRISPR/CasRx system to knockdown E2F8 mRNA, resulting in effective and specific downregulation of E2F8 and its target oncogenes, as well as significant growth inhibition in AR-negative CRPC in both cultured cells and xenograft models. Our findings identify and characterize E2F8 as a targetable transcriptional activator driving CRPC, particularly the growth of AR-negative CRPC.
Collapse
Affiliation(s)
- Furong Huang
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Kexin Li
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Zhong Chen
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Zhifen Cui
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - William Hankey
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Kun Fang
- Data Science Institute, MCW Cancer Center and Mellowes Center for Genome Science and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jingyue Yan
- Icahn Genomics Institute, Precision Immunology Institute, Department of Immunology and Immunotherapy, Department of Oncological Sciences, Tisch Cancer Institute, Biomedical Engineering and Imaging Institute, Friedman Brain Institute , Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hongyan Wang
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Victor X Jin
- Data Science Institute, MCW Cancer Center and Mellowes Center for Genome Science and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yizhou Dong
- Icahn Genomics Institute, Precision Immunology Institute, Department of Immunology and Immunotherapy, Department of Oncological Sciences, Tisch Cancer Institute, Biomedical Engineering and Imaging Institute, Friedman Brain Institute , Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Qianben Wang
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA.
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
24
|
Yang R, Fei L, Xue Y, Zhang Y, Hu Q, Guo L, Wei Y, Wu Q. Increasing RB1 Expression by Targeting EZH2 in Triple-Negative Breast Cancer. J Cell Mol Med 2025; 29:e70384. [PMID: 40070134 PMCID: PMC11897054 DOI: 10.1111/jcmm.70384] [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: 05/27/2024] [Revised: 01/06/2025] [Accepted: 01/15/2025] [Indexed: 03/15/2025] Open
Abstract
Loss of RB1 function represents a defining characteristic of triple-negative breast cancer (TNBC) and is intricately associated with resistance to therapeutic interventions. In this study, we investigate the epigenetic mechanisms governing RB1 expression in TNBC. Employing a combination of bioinformatics analyses and experimental validations, we identified lysine histone methyltransferase EZH2 as a key upstream regulator of RB1 expression. EZH2 primarily mediates trimethylation of lysine 27 on histone H3 as the catalytic subunit of the Polycomb repressive complex 2 (PRC2) complex. Furthermore, our findings demonstrate that pharmacological inhibition of EZH2 leads to a significant upregulation of RB1 expression levels, mediated by enhanced enrichment of the activating histone marker H3K27ac at the RB1 enhancer region, as evidenced by ATAC-sequencing and ChIP-qPCR assays. These insights unveil a promising clinical avenue for combating RB1-mediated drug resistance in TNBC through the strategic integration of epigenetic-targeting agents.
Collapse
Affiliation(s)
- Renfei Yang
- Hangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouChina
| | - Liyan Fei
- Hangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouChina
- School of Chemistry and Material ScienceUniversity of Science and Technology of ChinaHefeiChina
| | - Yingfei Xue
- Hangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouChina
- School of Pharmaceutical Science and Technology (SPST)Tianjin UniversityTianjinChina
| | - Yu Zhang
- Hangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouChina
- College of PharmacyZhejiang University of TechnologyHangzhouChina
| | - Qian Hu
- Hangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouChina
- Hangzhou Institute for Advanced Study (UCAS)Chinese Academy of SciencesHangzhouChina
| | - Lu Guo
- Hangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouChina
| | - Yong Wei
- Hangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouChina
| | - Qin Wu
- Hangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouChina
| |
Collapse
|
25
|
Tang H, Jiang X, Zhu L, Xu L, Wang X, Li H, Gao F, Liu X, Ren C, Zhao Y. Clinicopathologic and molecular characteristics of neuroendocrine carcinomas of the gallbladder. Histol Histopathol 2025; 40:389-400. [PMID: 39041213 DOI: 10.14670/hh-18-788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Gallbladder neuroendocrine carcinomas (GB-NECs) are a rare subtype of malignant gallbladder cancer (GBC). The genetic and molecular characteristics of GB-NECs are rarely reported. This study aims to assess the frequency of microsatellite instability (MSI) in GB-NECs and characterize their clinicopathologic and molecular features in comparison with gallbladder adenocarcinomas (GB-ADCs). Data from six patients with primary GB-NECs and 13 with GB-ADCs were collected and reevaluated. MSI assay, immunohistochemistry for mismatch repair proteins (MLH1, MSH2, MSH6, and PMS2), comprehensive genomic profiling (CGP) via next-generation sequencing (NGS), and evaluation of tumor mutation burden (TMB) were conducted on these samples. The six GB-NEC cases were all female, with a mean age of 62.0±9.2 years. Of these, two cases were diagnosed as large cell neuroendocrine carcinomas (LCNECs), while the remaining four were small cell neuroendocrine carcinomas (SCNECs). Microsatellite states observed in both GB-NECs and GB-ADCs were consistently microsatellite stable (MSS). Notably, TP53 (100%, 6/6) and RB1 (100%, 6/6) exhibited the highest mutation frequency in GB-NECs, followed by SMAD4 (50%, 3/6), GNAS (50%, 3/6), and RICTOR (33%, 2/6), with RB1, GNAS, and RICTOR specifically present in GB-NECs. Immunohistochemical (IHC) assays of p53 and Rb in the six GB-NECs were highly consistent with genetic mutations detected by targeted NGS. Moreover, no statistical difference was observed in TMB between GB-NECs and GB-ADCs (p=0.864). Although overall survival in GB-NEC patients tended to be worse than in GB-ADC patients, this difference did not reach statistical significance (p=0.119). This study has identified the microsatellite states and molecular mutation features of GB-NECs, suggesting that co-mutations in TP53 and RB1 may signify a neuroendocrine inclination in GB-NECs. The IHC assay provides an effective complement to targeted NGS for determining the functional status of p53 and Rb in clinical practice.
Collapse
Affiliation(s)
- Hui Tang
- Department of Pathology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaojun Jiang
- Department of Pathology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lili Zhu
- Department of Pathology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Liming Xu
- Department of Pathology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaoxi Wang
- Department of Pathology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hong Li
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Feifei Gao
- Department of Radiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xinxin Liu
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Chuanli Ren
- Department of Laboratory Medicine, Northern Jiangsu People's Hospital, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| | - Yan Zhao
- Medical Research Center, Northern Jiangsu People's Hospital, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China.
| |
Collapse
|
26
|
Ding CKC, Greenland NY, Sirohi D, Lotan TL. Molecular Landscape of Aggressive Histologic Subtypes of Localized Prostate Cancer. Surg Pathol Clin 2025; 18:1-12. [PMID: 39890297 DOI: 10.1016/j.path.2024.10.001] [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] [Indexed: 02/03/2025]
Abstract
Despite incredible progress in describing the molecular underpinnings of prostate cancer over the last decades, pathologic examination remains indispensable for predicting aggressive behavior in the localized setting. Beyond pathologic grade, specific histologic findings have emerged as critical prognostic or predictive indicators. Here, the authors review molecular correlates of aggressive histologic subtypes of prostate cancer in the localized setting, demonstrating that many of the signature molecular alterations found in metastatic disease-such as tumor suppressor gene loss and DNA repair defects-are enriched in primary disease with adverse histologic features, presaging aggressive behavior, and presenting opportunities for earlier germline screening or targeted therapies.
Collapse
Affiliation(s)
- Chien-Kuang C Ding
- Department of Pathology, University of California, San Francisco (UCSF), 1825 4th Street, M2370, San Francisco, CA 94158, USA
| | - Nancy Y Greenland
- Department of Pathology, University of California, San Francisco (UCSF), 1825 4th Street, M2370, San Francisco, CA 94158, USA
| | - Deepika Sirohi
- Department of Pathology, University of California, San Francisco (UCSF), 1825 4th Street, M2370, San Francisco, CA 94158, USA
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
27
|
Fukui T, Okasho K, Okuno Y, Fujiwara M, Hikami K, Fukunaga A, Sunada T, Kita Y, Sumiyoshi T, Goto T, Saito R, Ogawa O, Kobayashi T, Akamatsu S. A highly sensitive screening system to evaluate the reversibility of neuroendocrine prostate cancer to prostate adenocarcinoma. Cancer Med 2025; 14:e70047. [PMID: 40013333 PMCID: PMC11865886 DOI: 10.1002/cam4.70047] [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: 03/22/2024] [Revised: 05/23/2024] [Accepted: 07/12/2024] [Indexed: 02/28/2025] Open
Abstract
We established a robust and sensitive androgen response element luciferase reporter assay to monitor androgen receptor (AR) activity using KUCaP13 cells, a novel human-derived treatment-related neuroendocrine prostate cancer (t-NEPC) cell line. A high-throughput screening using a chemical library to identify potential compounds that induce AR re-expression in KUCaP13 cells revealed 30 candidate molecules potentially enhancing luciferase luminescence; however, subsequent validation steps demonstrated these signals to be false-positives. Despite not achieving the goal of AR re-expression, this study stands as a significant proof-of-concept for the application of high-throughput screening approaches in t-NEPC research.
Collapse
Affiliation(s)
- Tomohiro Fukui
- Department of UrologyKyoto University Graduate School of MedicineKyotoJapan
| | - Kosuke Okasho
- Department of UrologyKyoto University Graduate School of MedicineKyotoJapan
| | - Yukiko Okuno
- Medical Research Support Center, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Maki Fujiwara
- Department of UrologyKyoto University Graduate School of MedicineKyotoJapan
| | - Kensuke Hikami
- Department of UrologyKyoto University Graduate School of MedicineKyotoJapan
| | - Arinobu Fukunaga
- Department of UrologyKyoto University Graduate School of MedicineKyotoJapan
| | - Takuro Sunada
- Department of UrologyKyoto University Graduate School of MedicineKyotoJapan
| | - Yuki Kita
- Department of UrologyKyoto University Graduate School of MedicineKyotoJapan
| | - Takayuki Sumiyoshi
- Department of UrologyKyoto University Graduate School of MedicineKyotoJapan
| | - Takayuki Goto
- Department of UrologyKyoto University Graduate School of MedicineKyotoJapan
| | - Ryoichi Saito
- Department of UrologyKyoto University Graduate School of MedicineKyotoJapan
| | - Osamu Ogawa
- Department of UrologyKyoto University Graduate School of MedicineKyotoJapan
| | - Takashi Kobayashi
- Department of UrologyKyoto University Graduate School of MedicineKyotoJapan
| | - Shusuke Akamatsu
- Department of UrologyKyoto University Graduate School of MedicineKyotoJapan
- Department of UrologyNagoya University Graduate School of MedicineNagoyaJapan
| |
Collapse
|
28
|
Xue Y, Zhu L, Karan S, Locker JD, Branch C, Zhang J, Hoang B, Maianti JP, Zhao H, Schwartz EL. Distinct outcomes from targeted perturbations of the multi-subunit SCF Skp2 E3 ubiquitin ligase in blocking Trp53/Rb1-null prostate tumorigenesis. Commun Biol 2025; 8:278. [PMID: 39987265 PMCID: PMC11846996 DOI: 10.1038/s42003-025-07662-3] [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: 09/18/2023] [Accepted: 02/04/2025] [Indexed: 02/24/2025] Open
Abstract
Identifying effective therapies targeting multi-protein complexes that lack catalytic sites or cofactor pockets remains a long-standing challenge. The proto-oncogene, ubiquitin E3 ligase SCFSkp2, is one such target. SCFSkp2 promotes the proteasomal degradation of the cyclin-dependent kinase inhibitor p27, which controls cell cycle progression. Targeted knockout of Rb1/Trp53 causes metastatic prostate cancer in mice; additional knockout of Skp2 completely blocks tumorigenesis. We compared gene-edited mice that carried two different single amino acid changes in the SCFSkp2 complex, structurally predicted to inhibit the degradation of p27. Mutation of the SCFSkp2 accessory protein Cks1 (Cks1N45R) completely blocked Rb1/Trp53-driven prostate tumorigenesis, phenocopying Skp2 knockout, whereas a mutation directly stabilizing p27 (p27T187A) did not. This was consistent with structural models that predicted the binding of both p27 and p27T187A to the SCFSkp2/Cks1/Cdk2/CyclinA/p27 complex, and their subsequent ubiquitination and degradation, albeit at different rates. Two binding modes, which differ in their dependence on phosphorylated T187, are predicted by the model. Studies confirmed the role of p27 in mediating tumorigenesis in Rb1/Trp53 mutant tumors and revealed a mutually destabilizing Skp2 and p27 feedback loop. The integration of gene editing, drug-surrogate mutations, and mouse tumor models offers a blueprint for studying SCFSkp2 and other multi-subunit biomedical targets.
Collapse
Affiliation(s)
- Yingjiao Xue
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York, NY, USA
| | - Liang Zhu
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York, NY, USA
- Department of Ophthalmology & Visual Sciences, Albert Einstein College of Medicine, Bronx, New York, NY, USA
| | - Saumen Karan
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York, NY, USA
| | - Joseph D Locker
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Craig Branch
- Department of Radiology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jinghang Zhang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Bang Hoang
- Department of Orthopedic Surgery, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Juan Pablo Maianti
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Hongling Zhao
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York, NY, USA.
| | - Edward L Schwartz
- Department of Oncology, Albert Einstein College of Medicine, Bronx, NY, USA.
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
29
|
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
|
30
|
SONG S, WANG X, ZHOU S, CHENG X, LIN W, WANG Y, SUN Y. [Crosstalk between Tumor Cells and Neural Signals in Neuroendocrine Carcinoma
Metastasis: Communication Hijacking Based Perspective]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2025; 28:138-145. [PMID: 40114490 PMCID: PMC11931239 DOI: 10.3779/j.issn.1009-3419.2025.101.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Indexed: 03/22/2025]
Abstract
Neuroendocrine carcinoma (NEC) represents a category of malignant tumors originating from neuroendocrine cells. Given that NEC cells exhibit characteristics of both neural and endocrine cells, they can hijack neuronal signaling pathways and dynamically regulate the expression of neuronal lineage markers during tumor metastasis, thereby constructing a microenvironment conducive to tumor growth and metastasis. Conversely, alterations in the tumor microenvironment can enhance the interactions between neurons and tumor cells, ultimately synergistically promoting the metastasis of NEC. This review highlights recent advancements in the field of cancer neuroscience, uncovering neuronal lineage markers in NEC that facilitate tumor dissemination through mediating crosstalk, bidirectional communication, and synergistic interactions between tumor cells and the nervous system. Consequently, the latest findings in tumor neuroscience have enriched our understanding of the biological mechanisms underlying tumor metastasis, opening new research avenues for a deeper comprehension of the complex biological processes involved in tumor metastasis, particularly brain metastasis. This review provides a comprehensive review of the crosstalk between tumor cells and neural signaling in the metastasis of NEC.
.
Collapse
|
31
|
Liu S, Yu H, Zhao Z. The molecular pathogenesis of SOX2 in prostate cancer. Discov Oncol 2025; 16:215. [PMID: 39976818 PMCID: PMC11842661 DOI: 10.1007/s12672-025-01972-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/11/2025] [Indexed: 02/23/2025] Open
Abstract
SOX2 is one of the members of the SOX transcription factor family, which is believed to be an important transcription factor that plays a role in embryonic development, maintenance of stem cells, cancer progression, and resistance to cancer treatment. There is increasing evidence suggesting that SOX2 is crucial for the initiation, progression, invasion, metastasis, and treatment resistance of prostate cancer, therefore understanding the mechanism of SOX2 in prostate cancer can provide better targets for the treatment of prostate cancer. This article reviews the structural domains, normal physiological functions, and role in prostate cancer progression of SOX2, providing potential targets for prostate cancer treatment.
Collapse
Affiliation(s)
- Shixue Liu
- Jining Medical University, Jining, 272067, Shandong, China
| | - Honglian Yu
- Department of Biochemistry, Jining Medical University, 133 Hehua Road, Jining, 272067, Shandong, China.
| | - Zhankui Zhao
- Department of Urology, Affiliated Hospital of Jining Medical University, Jining Medical University, 89 Guhuai Road, Jining, 272029, Shandong, China.
- Department of Urology, Seventh People'S Hospital of Shanghai University of TCM, Shanghai, 200137, Shanghai, China.
| |
Collapse
|
32
|
Jiang Z, Allkanjari MS, Chung PED, Tran H, Ghanbari-Azarnier R, Wang DY, Lin DJ, Min JY, Ben-David Y, Zacksenhaus E. Recent Advances in Pineoblastoma Research: Molecular Classification, Modelling and Targetable Vulnerabilities. Cancers (Basel) 2025; 17:720. [PMID: 40075567 PMCID: PMC11898778 DOI: 10.3390/cancers17050720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/17/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
Pineoblastoma (PB) is a rare yet lethal pediatric brain cancer of the pineal gland, a small endocrine organ that secretes melatonin to regulate the circadian rhythm. For PB patients ≤5 years of age, the overall survival rate is approximately 15%; metastatic PB is incurable. Standard treatment, including surgical resection, radiation, and systemic chemotherapy, improves survival but compromises neurocognitive function. A better understanding of the disease and the generation of preclinical models may enable re-evaluation of previous clinical trials, development of precision therapeutic strategies and improve patient outcome. Over the past 5 years, PB has been recognized to include several major subtypes driven by (i) loss of microRNA processing factors DICER and DROSHA characterized by a relatively good prognosis; (ii) loss of the retinoblastoma tumor suppressor RB1; and (iii) amplification or induction of the cMYC protooncogene, with the latter two subtypes exhibiting exceedingly poor prognosis. Recently, mouse models for the major PB subtypes (RB1-, DICER1- and DROSHA-) except MYC- have been established. This progress, including better understanding of the disease, cell of origin, tumor progression, role of autophagy, and targetable vulnerabilities, holds promise for novel therapeutic strategies to combat each subtype of this lethal childhood malignancy.
Collapse
Affiliation(s)
- Zhe Jiang
- Toronto General Research Institute, University Health Network, 101 College Street, Max Bell Research Centre, Suite 5R406, Toronto, ON M5G 1L7, Canada; (M.S.A.); (H.T.); (R.G.-A.); (D.-Y.W.)
| | - Michelle S. Allkanjari
- Toronto General Research Institute, University Health Network, 101 College Street, Max Bell Research Centre, Suite 5R406, Toronto, ON M5G 1L7, Canada; (M.S.A.); (H.T.); (R.G.-A.); (D.-Y.W.)
| | - Philip E. D. Chung
- Toronto General Research Institute, University Health Network, 101 College Street, Max Bell Research Centre, Suite 5R406, Toronto, ON M5G 1L7, Canada; (M.S.A.); (H.T.); (R.G.-A.); (D.-Y.W.)
| | - Hanna Tran
- Toronto General Research Institute, University Health Network, 101 College Street, Max Bell Research Centre, Suite 5R406, Toronto, ON M5G 1L7, Canada; (M.S.A.); (H.T.); (R.G.-A.); (D.-Y.W.)
| | - Ronak Ghanbari-Azarnier
- Toronto General Research Institute, University Health Network, 101 College Street, Max Bell Research Centre, Suite 5R406, Toronto, ON M5G 1L7, Canada; (M.S.A.); (H.T.); (R.G.-A.); (D.-Y.W.)
| | - Dong-Yu Wang
- Toronto General Research Institute, University Health Network, 101 College Street, Max Bell Research Centre, Suite 5R406, Toronto, ON M5G 1L7, Canada; (M.S.A.); (H.T.); (R.G.-A.); (D.-Y.W.)
| | - Daniel J. Lin
- Toronto General Research Institute, University Health Network, 101 College Street, Max Bell Research Centre, Suite 5R406, Toronto, ON M5G 1L7, Canada; (M.S.A.); (H.T.); (R.G.-A.); (D.-Y.W.)
| | - Jung Yeon Min
- Toronto General Research Institute, University Health Network, 101 College Street, Max Bell Research Centre, Suite 5R406, Toronto, ON M5G 1L7, Canada; (M.S.A.); (H.T.); (R.G.-A.); (D.-Y.W.)
| | - Yaacov Ben-David
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang 550004, China
| | - Eldad Zacksenhaus
- Toronto General Research Institute, University Health Network, 101 College Street, Max Bell Research Centre, Suite 5R406, Toronto, ON M5G 1L7, Canada; (M.S.A.); (H.T.); (R.G.-A.); (D.-Y.W.)
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| |
Collapse
|
33
|
Sipola J, Munzur AD, Kwan EM, Seo CCY, Hauk BJ, Parekh K, Liao YJ(R, Bernales CQ, Donnellan G, Bloise I, Fung E, Ng SWS, Wang G, Vandekerkhove G, Nykter M, Annala M, Maurice-Dror C, Chi KN, Herberts C, Wyatt AW, Takeda DY. Plasma Cell-Free DNA Chromatin Immunoprecipitation Profiling Depicts Phenotypic and Clinical Heterogeneity in Advanced Prostate Cancer. Cancer Res 2025; 85:791-807. [PMID: 39652574 PMCID: PMC11832346 DOI: 10.1158/0008-5472.can-24-2052] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/21/2024] [Accepted: 12/04/2024] [Indexed: 02/18/2025]
Abstract
Cell phenotype underlies prostate cancer presentation and treatment resistance and can be regulated by epigenomic features. However, the osteotropic tendency of prostate cancer limits access to metastatic tissue, meaning most prior insights into prostate cancer chromatin biology are from preclinical models that do not fully represent disease complexity. Noninvasive chromatin immunoprecipitation of histones in plasma cell-free DNA (cfDNA) in humans may enable the capture of disparate prostate cancer phenotypes. In this study, we analyzed activating promoter- and enhancer-associated H3K4me2 from cfDNA in metastatic prostate cancer enriched for divergent patterns of metastasis and diverse clinical presentation. H3K4me2 density across prostate cancer genes, accessible chromatin, and lineage-defining transcription factor-binding sites correlated strongly with ctDNA fraction-demonstrating capture of prostate cancer-specific biology and informing the development of a statistical framework to adjust for ctDNA fraction. Chromatin hallmarks mirrored synchronously measured clinicogenomic features: bone- versus liver-predominant disease, serum PSA, biopsy-confirmed histopathologic subtype, and RB1 deletions convergently indicated phenotype segregation along an axis of differential androgen receptor activity and neuroendocrine identity. Detection of lineage switching after sequential progression on systemic therapy in select patients indicates potential use for individualized resistance monitoring. Epigenomic footprints of metastasis-induced normal tissue destruction were evident in bulk cfDNA from two patients. Finally, a public epigenomic resource was generated using a distinct chromatin marker that has not been widely investigated in prostate cancer. These results provide insights into the adaptive molecular landscape of aggressive prostate cancer and endorse plasma cfDNA chromatin profiling as a biomarker source and biological discovery tool. Significance: Plasma cell-free chromatin immunoprecipitation sequencing enables phenotypic dissection of lethal prostate cancer and is a practical tool for biomarker discovery while overcoming prior limitations of access to relevant tissue and reliance on model systems.
Collapse
Affiliation(s)
- Joonatan Sipola
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, Tampere, Finland
| | - Asli D. Munzur
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada
| | - Edmond M. Kwan
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada
- Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada
- Department of Medicine, School of Clinical Sciences; Monash University; Melbourne, Victoria, Australia
| | - Clara C. Y. Seo
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Benjamin J. Hauk
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Karan Parekh
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada
| | - Yi Jou (Ruby) Liao
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada
| | - Cecily Q. Bernales
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada
| | - Gráinne Donnellan
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada
| | - Ingrid Bloise
- Instituto Brasileiro de Controle ao Cancer, Sao Paulo, Brazil
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Emily Fung
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Sarah W. S. Ng
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada
| | - Gang Wang
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Gillian Vandekerkhove
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada
- Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada
| | - Matti Nykter
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, Tampere, Finland
| | - Matti Annala
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, Tampere, Finland
| | | | - Kim N. Chi
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada
- Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada
| | - Cameron Herberts
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada
| | - Alexander W. Wyatt
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, British Columbia, Canada
- Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - David Y. Takeda
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
34
|
Ofner H, Kramer G, Shariat SF, Hassler MR. TP53 Deficiency in the Natural History of Prostate Cancer. Cancers (Basel) 2025; 17:645. [PMID: 40002239 PMCID: PMC11853097 DOI: 10.3390/cancers17040645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/01/2025] [Accepted: 02/03/2025] [Indexed: 02/27/2025] Open
Abstract
Prostate cancer remains a leading cause of cancer-related mortality in men, with advanced stages posing significant treatment challenges due to high morbidity and mortality. Among genetic alterations, TP53 mutations are among the most prevalent in cancers and are strongly associated with poor clinical outcomes and therapeutic resistance. This review investigates the role of TP53 mutations in prostate cancer progression, prognosis, and therapeutic development. A comprehensive analysis of preclinical and clinical studies was conducted to elucidate the molecular mechanisms, clinical implications, and potential therapeutic approaches associated with TP53 alterations in prostate cancer. TP53 mutations are highly prevalent in advanced stages, contributing to genomic instability, aggressive tumor phenotypes, and resistance to standard treatments. Emerging evidence supports the utility of liquid biopsy techniques, such as circulating tumor DNA analysis, for detecting TP53 mutations, providing prognostic value and facilitating early intervention strategies. Novel therapeutic approaches targeting TP53 have shown promise in preclinical settings, but their clinical efficacy requires further validation. Overall, TP53 mutations represent a critical biomarker for disease progression and therapeutic response in prostate cancer. Advances in detection methods and targeted therapies hold significant potential to improve outcomes for patients with TP53-mutated prostate cancer. Further research is essential to integrate TP53-based strategies into routine clinical practice.
Collapse
Affiliation(s)
- Heidemarie Ofner
- Department of Urology, Medical University of Vienna, 1090 Vienna, Austria; (H.O.); (G.K.); (S.F.S.)
| | - Gero Kramer
- Department of Urology, Medical University of Vienna, 1090 Vienna, Austria; (H.O.); (G.K.); (S.F.S.)
| | - Shahrokh F. Shariat
- Department of Urology, Medical University of Vienna, 1090 Vienna, Austria; (H.O.); (G.K.); (S.F.S.)
- Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman 19328, Jordan
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Urology, Second Faculty of Medicine, Charles University, 150 06 Prague, Czech Republic
- Department of Urology, Weill Cornell Medical College, New York, NY 10065, USA
- Karl Landsteiner Institute of Urology and Andrology, 1090 Vienna, Austria
| | - Melanie R. Hassler
- Department of Urology, Medical University of Vienna, 1090 Vienna, Austria; (H.O.); (G.K.); (S.F.S.)
| |
Collapse
|
35
|
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
|
36
|
Gokbayrak B, Altintas UB, Lingadahalli S, Morova T, Huang CCF, Ersoy Fazlioglu B, Pak Lok Yu I, Kalkan BM, Cejas P, Kung SHY, Fazli L, Kawamura A, Long HW, Acilan C, Onder TT, Bagci-Onder T, Lynch JT, Lack NA. Identification of selective SWI/SNF dependencies in enzalutamide-resistant prostate cancer. Commun Biol 2025; 8:169. [PMID: 39905188 PMCID: PMC11794516 DOI: 10.1038/s42003-024-07413-w] [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: 02/28/2024] [Accepted: 12/17/2024] [Indexed: 02/06/2025] Open
Abstract
Enzalutamide is a potent second-generation antiandrogen commonly used to treat hormone-sensitive and castration-resistant prostate cancer (CRPC) patients. While initially effective, the disease almost always develops resistance. Given that many enzalutamide-resistant tumors lack specific somatic mutations, there is strong evidence that epigenetic factors can cause enzalutamide resistance. To explore how resistance arises, we systematically test all epigenetic modifiers in several models of castration-resistant and enzalutamide-resistant prostate cancer with a custom epigenetic CRISPR library. From this, we identify and validate SMARCC2, a core component of the SWI/SNF complex, that is selectivity essential in enzalutamide-resistant models. We show that the chromatin occupancy of SMARCC2 and BRG1 is expanded in enzalutamide resistance at regions that overlap with CRPC-associated transcription factors that are accessible in CRPC clinical samples. Overall, our study reveals a regulatory role for SMARCC2 in enzalutamide-resistant prostate cancer and supports the feasibility of targeting the SWI/SNF complex in late-stage PCa.
Collapse
Affiliation(s)
- Bengul Gokbayrak
- Koc University Research Centre for Translational Medicine (KUTTAM), Istanbul, Turkey
- Department of Clinical Pharmacology, School of Medicine, Koc University, Istanbul, Turkey
| | - Umut Berkay Altintas
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Shreyas Lingadahalli
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Tunc Morova
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Chia-Chi Flora Huang
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Betul Ersoy Fazlioglu
- Koc University Research Centre for Translational Medicine (KUTTAM), Istanbul, Turkey
- Department of Clinical Pharmacology, School of Medicine, Koc University, Istanbul, Turkey
| | - Ivan Pak Lok Yu
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Batuhan M Kalkan
- Koc University Research Centre for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Paloma Cejas
- Department of Medical Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, and Harvard Medical School, Boston, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, USA
- Translational Oncology Laboratory, Hospital La Paz Institute for Health Research (IdiPAZ) and CIBERONC, La Paz University Hospital, Madrid, Spain
| | - Sonia H Y Kung
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Ladan Fazli
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Akane Kawamura
- Chemistry - School of Natural and Environmental Sciences, Newcastle University, Newcastle, UK
- Department of Chemistry, University of Oxford, Oxford, UK
| | - Henry W Long
- Department of Medical Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, and Harvard Medical School, Boston, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, USA
| | - Ceyda Acilan
- Koc University Research Centre for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Tamer T Onder
- Koc University Research Centre for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Tugba Bagci-Onder
- Koc University Research Centre for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - James T Lynch
- Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Nathan A Lack
- Koc University Research Centre for Translational Medicine (KUTTAM), Istanbul, Turkey.
- Department of Clinical Pharmacology, School of Medicine, Koc University, Istanbul, Turkey.
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
37
|
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
|
38
|
Ding T, He L, Lin G, Xu L, Zhu Y, Wang X, Liu X, Guo J, Lei F, Zuo Z, Zheng J. Integrated analysis of single-cell and bulk transcriptomes uncovers clinically relevant molecular subtypes in human prostate cancer. Chin J Cancer Res 2025; 37:90-114. [PMID: 40078560 PMCID: PMC11893346 DOI: 10.21147/j.issn.1000-9604.2025.01.07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 12/26/2024] [Indexed: 03/14/2025] Open
Abstract
Objective Prostate cancer (PCa) is a complex disease characterized by diverse cellular ecosystems within the tumor microenvironment (TME) and high tumor heterogeneity, which challenges clinically stratified management and reinforces the need for novel strategies to fight against castration-resistant PCa (CRPC). Methods We performed single-cell RNA sequencing (scRNA-seq) on 10 untreated primary PCa tissues and integrated public scRNA-seq resources from three normal prostate tissues, two untreated primary PCa tissues, and six CRPC tumors to portray a comprehensive cellular and molecular interaction atlas of PCa. We further integrated the single-cell and bulk transcriptomes of PCa to establish a molecular classification system. Results scRNA-seq profiles revealed substantial inter- and intra-tumoral heterogeneity across different cell subpopulations in untreated PCa and CRPC tumors. In the malignant epithelial reservoir, cells evolved along decoupled paths in treatment-naive PCa and CRPC tumors, and distinct transcriptional reprogramming processes were activated, highlighting anti-androgen therapy-induced lineage plasticity. Based on the specifically expressed markers of the epithelial subpopulations, we conducted unsupervised clustering analysis in The Cancer Genome Atlas prostate adenocarcinoma (TCGA-PRAD) cohort and identified three molecularly and clinically distinct subtypes. The C1 subtype, characterized by high enrichment of CRPC-enriched epithelial cells, had a high risk of rapid development of anti-androgen resistance and might require active surveillance and additional promising intervention treatments, such as integrin A3 (ITGA3) + integrin B1 (ITGB1) inhibition. The C2 subtype resembled the immune-modulated subtype that was most likely to benefit from anti-LAG3 immunotherapy. The C3 subtype had a favorable prognosis. Conclusions Our study provides a comprehensive and high-resolution landscape of the intricate architecture of the PCa TME, and our trichotomic molecular taxonomy could help facilitate precision oncology.
Collapse
Affiliation(s)
- Tao Ding
- Department of Urology, Yuebei People’s Hospital, Shantou University Medical College, Shaoguan 512025, China
- Department of Urology, Southern Medical University Affifiliated Fengxian Hospital, Shanghai 201499, China
| | - Lina He
- School of Life Sciences, State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Guowen Lin
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Lei Xu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yanjun Zhu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xinan Wang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Xuefei Liu
- School of Life Sciences, State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Jianming Guo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Fanghong Lei
- Department of Pathology, Yuebei People’s Hospital, Shantou University Medical College, Shaoguan 512025, China
| | - Zhixiang Zuo
- School of Life Sciences, State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Jianghua Zheng
- Central Laboratory, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
- Department of Laboratory Medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| |
Collapse
|
39
|
Furlano K, Keshavarzian T, Biernath N, Fendler A, de Santis M, Weischenfeldt J, Lupien M. Epigenomics-guided precision oncology: Chromatin variants in prostate tumor evolution. Int J Cancer 2025. [PMID: 39853587 DOI: 10.1002/ijc.35327] [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: 09/09/2024] [Revised: 12/17/2024] [Accepted: 01/02/2025] [Indexed: 01/26/2025]
Abstract
Prostate cancer is a common malignancy that in 5%-30% leads to treatment-resistant and highly aggressive disease. Metastasis-potential and treatment-resistance is thought to rely on increased plasticity of the cancer cells-a mechanism whereby cancer cells alter their identity to adapt to changing environments or therapeutic pressures to create cellular heterogeneity. To understand the molecular basis of this plasticity, genomic studies have uncovered genetic variants to capture clonal heterogeneity of primary tumors and metastases. As cellular plasticity is largely driven by non-genetic events, complementary studies in cancer epigenomics are now being conducted to identify chromatin variants. These variants, defined as genomic loci in cancer cells that show changes in chromatin state due to the loss or gain of epigenomic marks, inclusive of histone post-translational modifications, DNA methylation and histone variants, are considered the fundamental units of epigenomic heterogeneity. In prostate cancer chromatin variants hold the promise of guiding the new era of precision oncology. In this review, we explore the role of epigenomic heterogeneity in prostate cancer, focusing on how chromatin variants contribute to tumor evolution and therapy resistance. We therefore discuss their impact on cellular plasticity and stochastic events, highlighting the value of single-cell sequencing and liquid biopsy epigenomic assays to uncover new therapeutic targets and biomarkers. Ultimately, this review aims to support a new era of precision oncology, utilizing insights from epigenomics to improve prostate cancer patient outcomes.
Collapse
Affiliation(s)
- Kira Furlano
- Department of Urology, Charité- Universitätsmedizin Berlin, Berlin, Germany
| | - Tina Keshavarzian
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Nadine Biernath
- Department of Urology, Charité- Universitätsmedizin Berlin, Berlin, Germany
| | - Annika Fendler
- Department of Urology, Charité- Universitätsmedizin Berlin, Berlin, Germany
| | - Maria de Santis
- Department of Urology, Charité- Universitätsmedizin Berlin, Berlin, Germany
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Joachim Weischenfeldt
- Department of Urology, Charité- Universitätsmedizin Berlin, Berlin, Germany
- Biotech Research & Innovation Centre (BRIC), The Finsen Laboratory, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Mathieu Lupien
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| |
Collapse
|
40
|
Wu F, Zhang H, Hao M. Interactions between key genes and pathways in prostate cancer progression and therapy resistance. Front Oncol 2025; 15:1467540. [PMID: 39917165 PMCID: PMC11799259 DOI: 10.3389/fonc.2025.1467540] [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: 07/20/2024] [Accepted: 01/06/2025] [Indexed: 02/09/2025] Open
Abstract
Prostate cancer is one of the most prevalent malignant tumors in men, particularly in regions with a high Human Development Index. While the long-term survival rate for localized prostate cancer is relatively high, the mortality rate remains significantly elevated once the disease progresses to advanced stages, even with various intensive treatment modalities. The primary obstacle to curing advanced prostate cancer is the absence of comprehensive treatment strategies that effectively target the highly heterogeneous tumors at both genetic and molecular levels. Prostate cancer development is a complex, multigenic, and multistep process that involves numerous gene mutations, alteration in gene expression, and changes in signaling pathways. Key genetic and pathway alterations include the amplification and/or mutation of the androgen receptor, the loss of Rb, PTEN, and p53, the activation of the WNT signaling pathway, and the amplification of the MYC oncogene. This review summarizes the mechanisms by which these genes influence the progression of prostate cancer and highlights the interactions between multiple genes and their relationship with prostate cancer. Additionally, we reviewed the current state of treatments targeting these genes and signaling pathways, providing a comprehensive overview of therapeutic approaches in the context of prostate cancer.
Collapse
Affiliation(s)
- Fan Wu
- Department of Pathology, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hengsen Zhang
- Department of Neurosurgery, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Miaomiao Hao
- Department of Pathology, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
41
|
Budreika A, Phoenix JT, Kostlan RJ, Deegan CD, Ferrari MG, Young KS, Fanning SW, Kregel S. The Homeobox Transcription Factor NKX3.1 Displays an Oncogenic Role in Castration-Resistant Prostate Cancer Cells. Cancers (Basel) 2025; 17:306. [PMID: 39858088 PMCID: PMC11763476 DOI: 10.3390/cancers17020306] [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: 12/10/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Prostate cancer (PCa) is the second leading cause of cancer-related death in men. The increase in incidence rates of more advanced and aggressive forms of the disease year-to-year fuels urgency to find new therapeutic interventions and bolster already established ones. PCa is a uniquely targetable disease in that it is fueled by male hormones (androgens) that drive tumorigenesis via the androgen receptor or AR. Current standard-of-care therapies directly target AR and its aberrant signaling axis but resistance to these therapies commonly arises, and the mechanisms behind the onset of therapy-resistance are still elusive. Research has shown that even with resistant disease, AR remains the main driver of growth and survival of PCa, and AR target genes and cofactors may help mediate resistance to therapy. Here, we focused on a homeobox transcription factor that exhibits a close relationship with AR-NKX3.1. Though NKX3.1 is traditionally thought of as a tumor suppressor, it has been previously reported to promote cancer cell survival by cooperating with AR. The role of NKX3.1 as a tumor suppressor perhaps in early-stage disease also contradicts its profile as a diagnostic biomarker for advanced prostate cancer. METHODS We investigated the physical interaction between NKX3.1 and AR, a modulated NKX3.1 expression in prostate cancer cells via overexpression and knockdown and assayed subsequent viability and downstream target gene expression. RESULTS We find that the expression of NKX3.1 is maintained in advanced PCa, and it is often elevated because of aberrant AR activity. Transient knockdown experiments across various PCa cell line models reveal NKX3.1 expression is necessary for survival. Similarly, stable overexpression of NKX3.1 in PCa cell lines reveals an androgen insensitive phenotype, suggesting NKX3.1 is sufficient to promote growth in the absence of an AR ligand. CONCLUSIONS Our work provides new insight into NKX3.1's oncogenic influence on PCa and the molecular interplay of these transcription factors in models of late-stage prostate cancer.
Collapse
Affiliation(s)
- Audris Budreika
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Stritch School of Medicine Health Sciences Division, Loyola University Chicago, 2160 South First Avenue Building 112, Room 205, Maywood, IL 60153, USA; (A.B.); (J.T.P.); (R.J.K.); (C.D.D.); (M.G.F.); (K.S.Y.); (S.W.F.)
- Integrated Program in Biomedical Science, Biochemistry, Molecular and Cancer Biology, Loyola University Chicago, Maywood, IL 60153, USA
| | - John T. Phoenix
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Stritch School of Medicine Health Sciences Division, Loyola University Chicago, 2160 South First Avenue Building 112, Room 205, Maywood, IL 60153, USA; (A.B.); (J.T.P.); (R.J.K.); (C.D.D.); (M.G.F.); (K.S.Y.); (S.W.F.)
- Integrated Program in Biomedical Science, Biochemistry, Molecular and Cancer Biology, Loyola University Chicago, Maywood, IL 60153, USA
| | - Raymond J. Kostlan
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Stritch School of Medicine Health Sciences Division, Loyola University Chicago, 2160 South First Avenue Building 112, Room 205, Maywood, IL 60153, USA; (A.B.); (J.T.P.); (R.J.K.); (C.D.D.); (M.G.F.); (K.S.Y.); (S.W.F.)
- Integrated Program in Biomedical Science, Biochemistry, Molecular and Cancer Biology, Loyola University Chicago, Maywood, IL 60153, USA
| | - Carleen D. Deegan
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Stritch School of Medicine Health Sciences Division, Loyola University Chicago, 2160 South First Avenue Building 112, Room 205, Maywood, IL 60153, USA; (A.B.); (J.T.P.); (R.J.K.); (C.D.D.); (M.G.F.); (K.S.Y.); (S.W.F.)
- Integrated Program in Biomedical Science, Biochemistry, Molecular and Cancer Biology, Loyola University Chicago, Maywood, IL 60153, USA
| | - Marina G. Ferrari
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Stritch School of Medicine Health Sciences Division, Loyola University Chicago, 2160 South First Avenue Building 112, Room 205, Maywood, IL 60153, USA; (A.B.); (J.T.P.); (R.J.K.); (C.D.D.); (M.G.F.); (K.S.Y.); (S.W.F.)
| | - Kristen S. Young
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Stritch School of Medicine Health Sciences Division, Loyola University Chicago, 2160 South First Avenue Building 112, Room 205, Maywood, IL 60153, USA; (A.B.); (J.T.P.); (R.J.K.); (C.D.D.); (M.G.F.); (K.S.Y.); (S.W.F.)
- Integrated Program in Biomedical Science, Biochemistry, Molecular and Cancer Biology, Loyola University Chicago, Maywood, IL 60153, USA
| | - Sean W. Fanning
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Stritch School of Medicine Health Sciences Division, Loyola University Chicago, 2160 South First Avenue Building 112, Room 205, Maywood, IL 60153, USA; (A.B.); (J.T.P.); (R.J.K.); (C.D.D.); (M.G.F.); (K.S.Y.); (S.W.F.)
| | - Steven Kregel
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Stritch School of Medicine Health Sciences Division, Loyola University Chicago, 2160 South First Avenue Building 112, Room 205, Maywood, IL 60153, USA; (A.B.); (J.T.P.); (R.J.K.); (C.D.D.); (M.G.F.); (K.S.Y.); (S.W.F.)
| |
Collapse
|
42
|
Joshi A, Bhaskar N, Pearson JD. Neuroendocrine Transformation as a Mechanism of Resistance to Targeted Lung Cancer Therapies: Emerging Mechanisms and Their Therapeutic Implications. Cancers (Basel) 2025; 17:260. [PMID: 39858043 PMCID: PMC11763869 DOI: 10.3390/cancers17020260] [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: 11/15/2024] [Revised: 12/24/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide, highlighting a major clinical challenge. Lung cancer is broadly classified into two histologically distinct subtypes, termed small cell lung cancer (SCLC) or non-small cell lung cancer (NSCLC). Identification of various oncogenic drivers of NSCLC has facilitated the development of targeted therapies that have dramatically improved patient outcomes. However, acquired resistance to these targeted therapies is common, which ultimately results in patient relapse. Several on-target and off-target resistance mechanisms have been described for targeted therapies in NSCLC. One common off-target mechanism of resistance to these therapies is histological transformation of the initial NSCLC into SCLC, a highly aggressive form of lung cancer that exhibits neuroendocrine histology. This mechanism of resistance presents a significant clinical challenge, since there are very few treatments available for these relapsed patients. Although the phenomenon of NSCLC-to-SCLC transformation was described almost 20 years ago, only recently have we begun to understand the mechanisms underlying this therapy-driven response. These recent discoveries will be key to identifying novel biomarkers and therapeutic strategies to improve outcomes of patients that undergo NSCLC-to-SCLC transformation. Here, we highlight these recent advances and discuss the potential therapeutic strategies that they have uncovered to target this mechanism of resistance.
Collapse
Affiliation(s)
- Asim Joshi
- Department of Pharmacology & Therapeutics, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3T 0T6, Canada; (A.J.); (N.B.)
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Nivitha Bhaskar
- Department of Pharmacology & Therapeutics, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3T 0T6, Canada; (A.J.); (N.B.)
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Joel D. Pearson
- Department of Pharmacology & Therapeutics, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3T 0T6, Canada; (A.J.); (N.B.)
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
- Children’s Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
| |
Collapse
|
43
|
Laisné M, Lupien M, Vallot C. Epigenomic heterogeneity as a source of tumour evolution. Nat Rev Cancer 2025; 25:7-26. [PMID: 39414948 DOI: 10.1038/s41568-024-00757-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/16/2024] [Indexed: 10/18/2024]
Abstract
In the past decade, remarkable progress in cancer medicine has been achieved by the development of treatments that target DNA sequence variants. However, a purely genetic approach to treatment selection is hampered by the fact that diverse cell states can emerge from the same genotype. In multicellular organisms, cell-state heterogeneity is driven by epigenetic processes that regulate DNA-based functions such as transcription; disruption of these processes is a hallmark of cancer that enables the emergence of defective cell states. Advances in single-cell technologies have unlocked our ability to quantify the epigenomic heterogeneity of tumours and understand its mechanisms, thereby transforming our appreciation of how epigenomic changes drive cancer evolution. This Review explores the idea that epigenomic heterogeneity and plasticity act as a reservoir of cell states and therefore as a source of tumour evolution. Best practices to quantify epigenomic heterogeneity and explore its various causes and consequences are discussed, including epigenomic reprogramming, stochastic changes and lasting memory. The design of new therapeutic approaches to restrict epigenomic heterogeneity, with the long-term objective of limiting cancer development and progression, is also addressed.
Collapse
Affiliation(s)
- Marthe Laisné
- CNRS UMR3244, Institut Curie, PSL University, Paris, France
- Translational Research Department, Institut Curie, PSL University, Paris, France
| | - Mathieu Lupien
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontorio, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, Ontorio, Canada.
- Ontario Institute for Cancer Research, Toronto, Ontorio, Canada.
| | - Céline Vallot
- CNRS UMR3244, Institut Curie, PSL University, Paris, France.
- Translational Research Department, Institut Curie, PSL University, Paris, France.
- Single Cell Initiative, Institut Curie, PSL University, Paris, France.
| |
Collapse
|
44
|
Pedrani M, Barizzi J, Salfi G, Nepote A, Testi I, Merler S, Castelo-Branco L, Mestre RP, Turco F, Tortola L, Theurillat JP, Gillessen S, Vogl U. The Emerging Predictive and Prognostic Role of Aggressive-Variant-Associated Tumor Suppressor Genes Across Prostate Cancer Stages. Int J Mol Sci 2025; 26:318. [PMID: 39796175 PMCID: PMC11719667 DOI: 10.3390/ijms26010318] [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: 12/03/2024] [Revised: 12/23/2024] [Accepted: 12/31/2024] [Indexed: 01/13/2025] Open
Abstract
Aggressive variant prostate cancer (AVPC) is characterized by a molecular signature involving combined defects in TP53, RB1, and/or PTEN (AVPC-TSGs), identifiable through immunohistochemistry or genomic analysis. The reported prevalence of AVPC-TSG alterations varies widely, reflecting differences in assay sensitivity, treatment pressure, and disease stage evolution. Although robust clinical evidence is still emerging, the study of AVPC-TSG alterations in prostate cancer (PCa) is promising. Alterations in TP53, RB1, and PTEN, as well as the combined loss of AVPC-TSGs, may have significant implications for prognosis and treatment. These biomarkers might help predict responses to various therapies, including hormonal treatments, cytotoxic agents, radiotherapy, and targeted therapies. Understanding the impact of these molecular alterations in patients with PCa is crucial for personalized management. In this review, we provide a comprehensive overview of the emerging prognostic and predictive roles of AVPC-TSG alterations across PCa stages. Moreover, we discuss the implications of different methods used for detecting AVPC-TSG alterations and summarize factors influencing their prevalence. As our comprehension of the genomic landscape of PCa disease deepens, incorporating genomic profiling into clinical decision making will become increasingly important for improving patient outcomes.
Collapse
Affiliation(s)
- Martino Pedrani
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.P.); (A.N.); (S.M.); (R.P.M.); (F.T.); (S.G.)
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, 20122 Milan, Italy
| | - Jessica Barizzi
- Istituto Cantonale di Patologia, Ente Ospedaliero Cantonale (EOC), 6600 Locarno, Switzerland
| | - Giuseppe Salfi
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.P.); (A.N.); (S.M.); (R.P.M.); (F.T.); (S.G.)
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland;
| | - Alessandro Nepote
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.P.); (A.N.); (S.M.); (R.P.M.); (F.T.); (S.G.)
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland;
- AOU San Luigi Gonzaga, Department of Oncology, University of Torino, 10124 Torino, Italy
| | - Irene Testi
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.P.); (A.N.); (S.M.); (R.P.M.); (F.T.); (S.G.)
- Medical Oncology Unit, University Hospital of Parma, 43126 Parma, Italy
| | - Sara Merler
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.P.); (A.N.); (S.M.); (R.P.M.); (F.T.); (S.G.)
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland;
- Section of Innovation Biomedicine—Oncology Area, Department of Engineering for Innovation Medicine, University of Verona and Verona University Hospital Trust, 37126 Verona, Italy
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| | - Luis Castelo-Branco
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.P.); (A.N.); (S.M.); (R.P.M.); (F.T.); (S.G.)
| | - Ricardo Pereira Mestre
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.P.); (A.N.); (S.M.); (R.P.M.); (F.T.); (S.G.)
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland;
| | - Fabio Turco
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.P.); (A.N.); (S.M.); (R.P.M.); (F.T.); (S.G.)
| | - Luigi Tortola
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.P.); (A.N.); (S.M.); (R.P.M.); (F.T.); (S.G.)
| | - Jean-Philippe Theurillat
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland;
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| | - Silke Gillessen
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.P.); (A.N.); (S.M.); (R.P.M.); (F.T.); (S.G.)
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| | - Ursula Vogl
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.P.); (A.N.); (S.M.); (R.P.M.); (F.T.); (S.G.)
| |
Collapse
|
45
|
Chen X, Wang K, Liao X, Zheng X, Yang S, Han C, Lu C, Wang X, Jin L, Kang H, Han Y, Wei J, Fan L, Zhang Z, Kong W. Single-Cell RNA Sequencing Reveals the Cellular Origin and Evolution of Small-Cell Neuroendocrine Carcinoma of the Cervix. J Med Virol 2025; 97:e70183. [PMID: 39831355 DOI: 10.1002/jmv.70183] [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: 08/14/2024] [Revised: 12/14/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025]
Abstract
Small-cell neuroendocrine cancer (SCNEC) of the uterine cervix is an exceedingly rare, highly aggressive tumor with an extremely poor prognosis. The cellular heterogeneity, origin, and tumorigenesis trajectories of SCNEC of the cervix remain largely unclear. We performed single-cell RNA sequencing and whole-exome sequencing on tumor tissues and adjacent normal cervical tissues from two patients diagnosed with SCNEC of the cervix. Here, we provide the first comprehensive insights into the cellular composition, HPV infection-related features, and gene expression profiles of SCNEC of the cervix at single-cell resolution. Correlation analyses suggested that SCNEC of the cervix may originate from squamous epithelial cells, and this observation was validated with bulk RNA-seq data from external cervical neuroendocrine cancer. Furthermore, sex-determining region Y-box 2 (SOX2), a key transcription factor that functions in direct neural differentiation, was located in the copy number gain region and highly expressed in neuroendocrine tumor cells from both patients. Notable, the distributions of the HPV-infected epithelium and SOX2 highly expressed epithelium were consistent with each other. Therefore, we supposed that high-risk HPV infection and amplification of SOX2 in the squamous epithelium may contribute to the progression of small-cell neuroendocrine tumorigenesis in the cervix.
Collapse
Affiliation(s)
- Xinyi Chen
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, P. R. China
| | - Kunyu Wang
- Department of Gynecology Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Xingyu Liao
- Familial & Hereditary Cancer Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, P. R. China
| | - Xingzheng Zheng
- Department of Pathology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, P. R. China
| | - Shuli Yang
- Department of Gynecology Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, P. R. China
| | - Chao Han
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, P. R. China
| | - Chang Lu
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, P. R. China
| | - Xiaodan Wang
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, P. R. China
| | - Lingge Jin
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, P. R. China
| | - Haili Kang
- Department of Gynecology Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, P. R. China
| | - Yiding Han
- Department of Pathology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, P. R. China
| | - Jiacong Wei
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Linyuan Fan
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, P. R. China
| | - Zhan Zhang
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, P. R. China
| | - Weimin Kong
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, P. R. China
| |
Collapse
|
46
|
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
|
47
|
Chen W, Mao Y, Zhan Y, Li W, Wu J, Mao X, Xu B, Shu F. Exosome-delivered NR2F1-AS1 and NR2F1 drive phenotypic transition from dormancy to proliferation in treatment-resistant prostate cancer via stabilizing hormonal receptors. J Nanobiotechnology 2024; 22:761. [PMID: 39695778 DOI: 10.1186/s12951-024-03025-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 11/19/2024] [Indexed: 12/20/2024] Open
Abstract
Cancer cells acquire the ability to reprogram their phenotype in response to targeted therapies, yet the transition from dormancy to proliferation in drug-resistant cancers remains poorly understood. In prostate cancer, we utilized high-plasticity mouse models and enzalutamide-resistant (ENZ-R) cellular models to elucidate NR2F1 as a key factor in lineage transition and ENZ resistance. Depletion of NR2F1 drives ENZ-R cells into a relative dormancy state, characterized by reduced proliferation and heightened drug resistance, while NR2F1 overexpression yields contrasting outcomes. Transcriptional sequencing analysis of NR2F1-silenced prostate cancer cells and tissues from the Cancer Genome Atlas-prostate cancer and SU2C cohorts indicated exosomes as the most enriched cell component, with pathways implicated in steroid hormone biosynthesis and drug metabolism. Moreover, NR2F1-AS1 forms a complex with SRSF1 to upregulate NR2F1 expression, facilitating its binding with ESR1 to sustain hormonal receptor expression and enhance proliferation in ENZ-R cells. Furthermore, HnRNPA2B1 interacts with NR2F1 and NR2F1-AS1, assisting their packaging into exosomes, wherein exosomal NR2F1 and NR2F1-AS1 promote the proliferation of dormant ENZ-R cells. Our works offer novel insights into the reawaking of dormant drug-resistant cancer cells governed by NR2F1 upregulation triggered by exosome-derived NR2F1-AS1 and NR2F1, suggesting therapeutic potential for phenotype reversal.
Collapse
Affiliation(s)
- Wenbin Chen
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Yiyou Mao
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - YiYuan Zhan
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenfeng Li
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Jun Wu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiangming Mao
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Bin Xu
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Fangpeng Shu
- Department of Urology, Guangzhou Women and Children's Medical Center, National Children's Medical Center for South Central Region, Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
48
|
Deng M, Huang PZ, Huang ZY, Chen TT, Luo X, Liao CY, Xu WH, Zhao J, Wu QJ, Zheng J. SOX2 control activation of dormant prostate cancer cells in bone metastases by promoting CCNE2 gene expression. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2024; 12:375-388. [PMID: 39839747 PMCID: PMC11744349 DOI: 10.62347/ascy2532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 12/12/2024] [Indexed: 01/23/2025]
Abstract
BACKGROUND Cancer stem cells (CSCs) have a powerful tumor initiation ability, which can promote the early dissemination of single disseminated tumor cells (DTCs), leading to tumor progression. SOX2, a pluripotent inducible transcription factor, is key to maintaining self-renewal and pluripotency of prostate cancer stem cells. However, there is a lack of comprehensive understanding of how SOX2 regulates DTCs dormancy and proliferation in the bone marrow microenvironment. METHODS AND RESULTS By constructing a mouse bone metastasis model to simulate the progression of prostate cancer with bone metastasis, the bone tissue immunofluorescence showed that SOX2 expression increased with the progression of prostate cancer in the bone marrow microenvironment. We validated this phenomenon with publicly available single-cell and transcriptome datasets and found that SOX2 is involved in multiple phenotypes associated with prostate cancer dormancy, proliferation, and invasion. Further, CCNE2, a potential target downstream of SOX2, was identified through multiple transcription factor databases and protein interaction networks. CONCLUSION The expression of SOX2 affects multiple phenotypes related to dormancy, proliferation and invasion of prostate cancer, and may indirectly activate the dormant prostate cancer cells through the downstream target gene CCNE2, thus affecting the progression and bone metastasis of prostate cancer.
Collapse
Affiliation(s)
- Min Deng
- Department of Urology, The Second Affiliated Hospital, Army Military Medical UniversityChongqing, China
| | - Pei-Zheng Huang
- Department of Urology, The Second Affiliated Hospital, Army Military Medical UniversityChongqing, China
- School of Medicine, Chongqing UniversityChongqing, China
| | - Ze-Yu Huang
- Department of Urology, The Second Affiliated Hospital, Army Military Medical UniversityChongqing, China
| | - Ting-Ting Chen
- Department of Urology, The Second Affiliated Hospital, Army Military Medical UniversityChongqing, China
| | - Xing Luo
- Department of Urology, The Second Affiliated Hospital, Army Military Medical UniversityChongqing, China
| | - Chao-Yu Liao
- Department of Urology, The Second Affiliated Hospital, Army Military Medical UniversityChongqing, China
| | - Wen-Hao Xu
- Department of Urology, The Second Affiliated Hospital, Army Military Medical UniversityChongqing, China
| | - Jiang Zhao
- Department of Urology, The Second Affiliated Hospital, Army Military Medical UniversityChongqing, China
| | - Qing-Jian Wu
- Department of Urology, The Second Affiliated Hospital, Army Military Medical UniversityChongqing, China
| | - Ji Zheng
- Department of Urology, The Second Affiliated Hospital, Army Military Medical UniversityChongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Third Military Medical UniversityChongqing, China
| |
Collapse
|
49
|
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
|
50
|
Varuzhanyan G, Chen CC, Freeland J, He T, Tran W, Song K, Wang L, Cheng D, Xu S, Dibernardo GA, Esedebe FN, Bhatia V, Han M, Abt ER, Park JW, Memarzadeh S, Shackelford DB, Lee JK, Graeber TG, Shirihai OS, Witte ON. PGC-1α drives small cell neuroendocrine cancer progression toward an ASCL1-expressing subtype with increased mitochondrial capacity. Proc Natl Acad Sci U S A 2024; 121:e2416882121. [PMID: 39589879 PMCID: PMC11626175 DOI: 10.1073/pnas.2416882121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 10/25/2024] [Indexed: 11/28/2024] Open
Abstract
Adenocarcinomas from multiple tissues can converge to treatment-resistant small cell neuroendocrine (SCN) cancers composed of ASCL1, POU2F3, NEUROD1, and YAP1 subtypes. We investigated how mitochondrial metabolism influences SCN cancer (SCNC) progression. Extensive bioinformatics analyses encompassing thousands of patient tumors and human cancer cell lines uncovered enhanced expression of proliferator-activatedreceptor gamma coactivator 1-alpha (PGC-1α), a potent regulator of mitochondrial oxidative phosphorylation (OXPHOS), across several SCNCs. PGC-1α correlated tightly with increased expression of the lineage marker Achaete-scute homolog 1, (ASCL1) through a positive feedback mechanism. Analyses using a human prostate tissue-based SCN transformation system showed that the ASCL1 subtype has heightened PGC-1α expression and OXPHOS activity. PGC-1α inhibition diminished OXPHOS, reduced SCNC cell proliferation, and blocked SCN prostate tumor formation. Conversely, PGC-1α overexpression enhanced OXPHOS, validated by small-animal Positron Emission Tomography mitochondrial imaging, tripled the SCN prostate tumor formation rate, and promoted commitment to the ASCL1 lineage. These results establish PGC-1α as a driver of SCNC progression and subtype determination, highlighting metabolic vulnerabilities in SCNCs across different tissues.
Collapse
Affiliation(s)
- Grigor Varuzhanyan
- Department of Microbiology Immunology and Molecular Genetics, University of California, Los Angeles, CA90095
| | - Chia-Chun Chen
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA90095
| | - Jack Freeland
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA90095
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, CA90095
| | - Tian He
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA90095
| | - Wendy Tran
- Department of Microbiology Immunology and Molecular Genetics, University of California, Los Angeles, CA90095
| | - Kai Song
- Department of Bioengineering, University of California, Los Angeles, CA90095
| | - Liang Wang
- Department of Microbiology Immunology and Molecular Genetics, University of California, Los Angeles, CA90095
| | - Donghui Cheng
- Department of Microbiology Immunology and Molecular Genetics, University of California, Los Angeles, CA90095
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA90095
| | - Shili Xu
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA90095
- Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Jonsson Comprehensive Cancer Center, the David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Gabriella A. Dibernardo
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA90095
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Favour N. Esedebe
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA90095
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, CA90095
| | - Vipul Bhatia
- Division of Hematology/Oncology, Department of Medicine University of California Los Angeles Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA90095
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA98109
| | - Mingqi Han
- Department of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Evan R. Abt
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA90095
| | - Jung Wook Park
- Department of Pathology, Duke University School of Medicine, Durham, NC27710
| | - Sanaz Memarzadeh
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA90095
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA90095
- Jonsson Comprehensive Cancer Center, the David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA90095
- The Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA90073
- Molecular Biology Institute, University of California, Los Angeles, CA90095
| | - David B. Shackelford
- Jonsson Comprehensive Cancer Center, the David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Department of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - John K. Lee
- Division of Hematology/Oncology, Department of Medicine University of California Los Angeles Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA90095
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA98109
| | - Thomas G. Graeber
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA90095
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA90095
- Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Jonsson Comprehensive Cancer Center, the David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, CA90095
- California NanoSystems Institute, University of California, Los Angeles, CA90095
- UCLA Metabolomics Center, University of California, Los Angeles, CA90095
| | - Orian S. Shirihai
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA90095
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, CA90095
- University of California Los Angeles Division of Endocrinology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Department of Clinical Biochemistry, School of Medicine, Ben Gurion University of The Negev, Beer-Sheva8410501, Israel
| | - Owen N. Witte
- Department of Microbiology Immunology and Molecular Genetics, University of California, Los Angeles, CA90095
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA90095
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA90095
- Jonsson Comprehensive Cancer Center, the David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Molecular Biology Institute, University of California, Los Angeles, CA90095
- Parker Institute for Cancer Immunotherapy, University of California, Los Angeles, CA90095
| |
Collapse
|