1
|
Song Y, Zhang K, Zhang J, Li Q, Huang N, Ma Y, Hou N, Han F, Kan C, Sun X. Epigenetic regulation of nuclear receptors: Implications for endocrine-related diseases and therapeutic strategies. Genes Dis 2025; 12:101481. [PMID: 40290121 PMCID: PMC12022648 DOI: 10.1016/j.gendis.2024.101481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/20/2024] [Accepted: 11/03/2024] [Indexed: 04/30/2025] Open
Abstract
The expression and function of the receptor are controlled by epigenetic changes, such as DNA methylation, histone modification, and noncoding RNAs. These modifications play a pivotal role in receptor activity and can lead to or exacerbate endocrine-related diseases. This review examines the epigenetic alterations of nuclear receptors and their significant impact on conditions such as diabetes, thyroid disorders, and endocrine-related tumors. It highlights current therapies targeting these epigenetic mechanisms, including gene editing, epigenetic drugs, and various other therapeutic approaches. This review offers fresh insight into the mechanisms of endocrine-associated disorders, highlighting the latest progress in the development of novel epigenetic therapies that can be used to address receptor-endocrine interactions.
Collapse
Affiliation(s)
- Yixin Song
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Kexin Zhang
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Jingwen Zhang
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Qinying Li
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Na Huang
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Yujie Ma
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Fang Han
- Department of Pathology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| |
Collapse
|
2
|
Abida W, Beltran H, Raychaudhuri R. State of the Art: Personalizing Treatment for Patients With Metastatic Castration-Resistant Prostate Cancer. Am Soc Clin Oncol Educ Book 2025; 45:e473636. [PMID: 40112242 DOI: 10.1200/edbk-25-473636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Until recently, the treatment of metastatic castration-resistant prostate cancer (mCRPC) relied exclusively on hormonal therapies and taxane chemotherapy. The advent of modern molecular profiling methods applied in the clinic, namely, next-generation sequencing and advanced positron emission tomography (PET) imaging, has allowed for the development of biomarker-driven therapeutics including anti-PD-L1 therapy for microsatellite instability-high or tumor mutation burden-high disease, poly(ADP-ribose) polymerase (PARP) inhibitors for patients with DNA damage repair mutations, and lutetium 177 vipivotide tetraxetan (177Lu-PSMA-617) for patients with prostate-specific membrane antigen (PSMA) PET-avid disease. While these targeted therapies have improved outcomes, there is an opportunity to refine biomarkers to optimize patient selection, understand resistance, and develop novel combination strategies. In addition, studies in the laboratory and in patient-derived samples have shown that a subset of mCRPC tumors lose expression of common prostate cancer markers such as prostate-specific antigen and PSMA because of lineage plasticity and the development of non-androgen receptor (AR)-driven disease. Non-AR-driven prostate cancer has been associated with aggressive behavior and poor prognosis, including in some cases histologic transformation to a poorly differentiated neuroendocrine prostate cancer (NEPC). The clinical management of NEPC typically follows the treatment paradigm for small cell lung cancer and increasingly relies on genomic and phenotypic characterization of disease, including loss of tumor suppressors and expression of cell surface markers such as DLL3. Therefore, both genomic subtyping and phenotypic subtyping are important to consider and can guide the clinical management of patients with advanced prostate cancer.
Collapse
Affiliation(s)
- Wassim Abida
- Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Ruben Raychaudhuri
- University of Washington and the Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
3
|
Cui Z, Huang F, Fang K, Yan J, Zhang Y, Kang DD, Zhou Y, Zhao Y, Everitt JI, Hankey W, Armstrong AJ, Huang J, Wang H, Jin VX, Dong Y, Wang Q. SCORT-Cas13d Nanotherapy Precisely Targets the 'Undruggable' Transcription Factor HoxB13 in Metastatic Prostate Cancer In Vivo. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2417605. [PMID: 40349174 DOI: 10.1002/advs.202417605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 04/01/2025] [Indexed: 05/14/2025]
Abstract
Metastatic cancer, the primary cause of cancer mortality, frequently exhibits heightened dependence on certain transcription factors (TFs), which serve as master regulators of oncogenic signaling yet are often untargetable by small molecules. Selective Cell in ORgan Targeting (SCORT) nanoparticles are developed for precise CRISPR/Cas13d mRNA and gRNA delivery to metastatic cancer cells in vivo, aiming to knock down the undruggable oncogenic TF HoxB13. In prostate cancer liver metastasis models driven by HoxB13, repeated systemic SCORT-Cas13d-gHoxB13 treatment significantly decreases HoxB13 expression, reduces metastasis, and extends mouse survival. Prolonged treatment shows no significant impact on major organ function, histology or immune markers. Mechanistically, SCORT-Cas13d-gHoxB13 treatment suppresses metastatic tumor proliferation and angiogenesis while promoting apoptosis by regulating multiple gene pathways. Unexpectedly, it inhibits the non-canonical, EMT-independent oncogenic function of Snail. These findings suggest that SCORT-Cas13d-gHoxB13 can effectively and safely target the undruggable HoxB13 in metastatic prostate cancer, positioning CRISPR/Cas13d as a potential treatment.
Collapse
Affiliation(s)
- Zhifen Cui
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Furong Huang
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Kun Fang
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Jingyue Yan
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Yuebao Zhang
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Diana D Kang
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
- 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, 10029, USA
| | - Yufan Zhou
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Yue Zhao
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Jeffrey I Everitt
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - William Hankey
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Andrew J Armstrong
- Department of Medicine, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Cancer Institute Center for Prostate and Urologic Cancer, Durham, NC, 27710, USA
| | - Jiaoti Huang
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Hongyan Wang
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Victor X Jin
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, 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, 10029, USA
| | - Qianben Wang
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Cancer Institute Center for Prostate and Urologic Cancer, Durham, NC, 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27710, USA
| |
Collapse
|
4
|
Walke P, Price JDW, Vizeacoumar FS, Joseph N, Maranda V, Chowdhury B, Patel J, Zhang Y, Dong H, New L, Ganapathysamy A, Gong LH, Lazell-Wright M, Elhasasna H, Bhanumathy KK, Wu Y, Wang Y, Freywald A, Krishnan A, Vizeacoumar FJ. A novel role for Neurog2 in MYCN driven neuroendocrine plasticity of prostate cancer. Oncogene 2025:10.1038/s41388-025-03413-0. [PMID: 40301542 DOI: 10.1038/s41388-025-03413-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 05/01/2025]
Abstract
Neuroendocrine prostate cancer (NEPC) presents a formidable clinical challenge owing to its aggressive progression and resistance to conventional therapies. A key driver of NEPC is the overexpression of MYCN, a well-established oncogene associated with neuroendocrine tumors. However, efforts to directly inhibit the N-Myc protein encoded by this gene have resulted in limited success, thereby hindering therapeutic advancements. To overcome this obstacle, we conducted unbiased genome-wide screening using isogenic prostate cancer cell lines to identify the synthetic vulnerabilities of MYCN. Among the identified candidates, NEUROG2 emerged as a significant candidate. Neurog2 is a proneural transcription factor (PTF) known for its role in developmental processes and trans-differentiation of adult cells. Our findings demonstrate that Neurog2 depletion does not affect non-malignant cells but significantly suppresses the growth of MYCN-overexpressing cells and tumors in orthotopic NEPC models. Furthermore, our observations indicate that Neurog2-driven modulation of PTFs potentially contribute to NEPC development. Thus, targeting Neurog2 holds promise as an effective therapeutic strategy for MYCN-overexpressing NEPC.
Collapse
Affiliation(s)
- Prachi Walke
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, and Cameco MS Neuroscience Research Centre, 701 Queen St, Saskatoon, S7K 0M7, SK, Canada
| | - Jared D W Price
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
| | - Frederick S Vizeacoumar
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
| | - Nickson Joseph
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, and Cameco MS Neuroscience Research Centre, 701 Queen St, Saskatoon, S7K 0M7, SK, Canada
| | - Vincent Maranda
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
| | - Bari Chowdhury
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, and Cameco MS Neuroscience Research Centre, 701 Queen St, Saskatoon, S7K 0M7, SK, Canada
| | - Jay Patel
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, and Cameco MS Neuroscience Research Centre, 701 Queen St, Saskatoon, S7K 0M7, SK, Canada
| | - Yue Zhang
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
| | - He Dong
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
| | - Lara New
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, and Cameco MS Neuroscience Research Centre, 701 Queen St, Saskatoon, S7K 0M7, SK, Canada
| | | | - Li Hui Gong
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
| | - Mary Lazell-Wright
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
| | - Hussain Elhasasna
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
| | - Kalpana K Bhanumathy
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
| | - Yuliang Wu
- Department of Biochemistry Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
| | - Yuzhuo Wang
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada.
- BC Cancer Research Centre, Vancouver, BC, Canada.
| | - Andrew Freywald
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada.
| | - Anand Krishnan
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, and Cameco MS Neuroscience Research Centre, 701 Queen St, Saskatoon, S7K 0M7, SK, Canada.
| | - Franco J Vizeacoumar
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada.
- Cancer Research Department, Saskatchewan Cancer Agency, 107 Wiggins Road, Saskatoon, S7N 5E5, SK, Canada.
| |
Collapse
|
5
|
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
|
6
|
Lin G, Elkashif A, Saha C, Coulter JA, Dunne NJ, McCarthy HO. Key considerations for a prostate cancer mRNA vaccine. Crit Rev Oncol Hematol 2025; 208:104643. [PMID: 39900315 DOI: 10.1016/j.critrevonc.2025.104643] [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: 12/20/2024] [Revised: 01/20/2025] [Accepted: 01/30/2025] [Indexed: 02/05/2025] Open
Abstract
Prostate cancer has the second highest cancer mortality rate in the UK in males. Early prostate cancer is typically asymptomatic, with diagnosis at a locally advanced or metastatic stage. In addition, the inherent heterogeneity of prostate cancer tumours differs significantly in terms of genetic, molecular, and histological features. The successful treatment of prostate cancer is therefore exceedingly challenging. Immunotherapies, particularly therapeutic vaccines, have been widely used in preclinical and clinical studies to treat various cancers. Sipuleucel-T was the first cancer vaccine approved by the FDA for the treatment of asymptomatic or minimally symptomatic metastatic castration-resistant prostate cancer (mCRPC), ushering in a new era of immunotherapy. In this review, the latest immunotherapy strategies for prostate cancer are considered with key tumour-associated antigens (TAA) and tumour-specific antigens (TSA) highlighted. The key components of mRNA vaccines include in vitro transcription, stability, and immunogenicity. Finally, strategies to circumvent in vivo mRNA degradation and approaches to optimise in vitro transcription (IVT) process are also discussed.
Collapse
Affiliation(s)
- Guanjie Lin
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Ahmed Elkashif
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Chayanika Saha
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Jonathan A Coulter
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Nicholas J Dunne
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin D09 NA55, Ireland; Centre for Medical Engineering Research, Dublin City University, Dublin D09 NA55, Ireland; Biodesign Europe, Dublin City University, Dublin D09 NA55, Ireland; Tissue, Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin D02 PN40, Ireland; Advanced Manufacturing Research Centre (I-Form), School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin D09 NA55, Ireland; Advanced Processing Technology Research Centre, Dublin City University, Dublin D09 NA55, Ireland; Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin D02 PN40, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin, Dublin D02 PN40, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin D02 PN40, Ireland
| | - Helen O McCarthy
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK.
| |
Collapse
|
7
|
Chiu CL, Zhang D, Zhao H, Wei Y, Polasko AL, Thomsen MT, Yang V, Yang KK, Hauck S, Peterson EE, Wen RM, Qiu Z, Corey E, Miao YR, Rankin EB, Peehl DM, Huang J, Giaccia AJ, Brooks JD. Targeting AXL Inhibits the Growth and Metastasis of Prostate Cancer in Bone. Clin Cancer Res 2025; 31:1346-1358. [PMID: 39879384 PMCID: PMC11961319 DOI: 10.1158/1078-0432.ccr-24-3028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/22/2024] [Accepted: 01/27/2025] [Indexed: 01/31/2025]
Abstract
PURPOSE After failing primary and secondary hormonal therapy, castration-resistant and neuroendocrine prostate cancer metastatic to the bone is invariably lethal, although treatment with docetaxel and carboplatin can modestly improve survival. Therefore, agents targeting biologically relevant pathways in prostate cancer and potentially synergizing with docetaxel and carboplatin in inhibiting bone metastasis growth are urgently needed. EXPERIMENTAL DESIGN Phosphorylated (activated) AXL expression in human prostate cancer bone metastases was assessed by IHC staining. We evaluated the effects of a novel soluble AXL signaling inhibitor, sAXL (batiraxcept or AVB-S6-500), on tumor growth and lung metastases in prostate cancer patient-derived xenograft models that were implanted intratibially. After injection of LuCaP cells into the tibiae, tumors were treated with batiraxcept and docetaxel or carboplatin alone or in combination, and tumor growth was monitored by serum prostate-specific antigen or bioluminescence. Tumor burden was quantified by human-specific Ku70 staining, and metastasis to the lungs was determined using qPCR. Transcriptomic profiling, Western blotting, and immunohistochemistry were performed to identify treatment-regulated gene and protein profile changes. RESULTS High AXL phosphorylation in human prostate cancer bone metastases correlated with shortened survival. Batiraxcept alone or in combination with docetaxel or carboplatin significantly suppressed intratibial tumor growth and suppressed metastasis to the lungs through multiple mechanisms, including repression of cancer stemness genes (CD44, ALDH1A1, TACSTD2, and ATXN1) and the PI3K, JAK, MAPK, and E2F1/NUSAP1 signaling pathways. CONCLUSIONS Our study provides a robust preclinical rationale and mechanisms of action for using batiraxcept as a single agent or in combination with docetaxel or carboplatin to treat lethal metastatic prostate cancer.
Collapse
Affiliation(s)
- Chun-Lung Chiu
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Dalin Zhang
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Hongjuan Zhao
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yi Wei
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Mikkel Thy Thomsen
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Clinical Medicine, Aarhus University, Denmark
| | - Vanessa Yang
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kasie Kexin Yang
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Spencer Hauck
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Eric E. Peterson
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ru M. Wen
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Zhengyuan Qiu
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, Washington, USA
| | - Yu Rebecca Miao
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Erinn B. Rankin
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Donna M. Peehl
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Jiaoti Huang
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Amato J. Giaccia
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Oncology, University of Oxford, Oxford, UK
| | - James D. Brooks
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Research Institute, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Hofstad M, Woods A, Parra K, Sychev ZE, Mazzagatti A, Huo X, Yu L, Gilbreath C, Chen WM, Davis AJ, Ly P, Drake JM, Kittler R. Dual inhibition of ATR and DNA-PKcs radiosensitizes ATM-mutant prostate cancer. Oncogene 2025:10.1038/s41388-025-03343-x. [PMID: 40119228 DOI: 10.1038/s41388-025-03343-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 01/31/2025] [Accepted: 03/06/2025] [Indexed: 03/24/2025]
Abstract
In advanced castration resistant prostate cancer (CRPC), mutations in the DNA damage response (DDR) gene ataxia telangiectasia mutated (ATM) are common. While poly(ADP-ribose) polymerase inhibitors are approved in this context, their clinical efficacy remains limited. Thus, there is a compelling need to identify alternative therapeutic avenues for ATM mutant prostate cancer patients. Here, we generated matched ATM-proficient and ATM-deficient CRPC lines to elucidate the impact of ATM loss on DDR in response to DNA damage via irradiation. Through unbiased phosphoproteomic screening, we unveiled that ATM-deficient CRPC lines maintain dependence on downstream ATM targets through activation of ATR and DNA-PKcs kinases. Dual inhibition of ATR and DNA-PKcs effectively inhibited downstream γH2AX foci formation in response to irradiation and radiosensitized ATM-deficient lines to a greater extent than either ATM-proficient controls or single drug treatment. Further, dual inhibition abrogated residual downstream ATM pathway signaling and impaired replication fork dynamics. To circumvent potential toxicity, we leveraged the RUVBL1/2 ATPase inhibitor Compound B, which leads to the degradation of both ATR and DNA-PKcs kinases. Compound B effectively radiosensitized ATM-deficient CRPC in vitro and in vivo, and impacted replication fork dynamics. Overall, dual targeting of both ATR and DNA-PKcs is necessary to block DDR in ATM-deficient CRPC, and Compound B could be utilized as a novel therapy in combination with irradiation in these patients.
Collapse
Affiliation(s)
- Mia Hofstad
- Eugene McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, USA.
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Andrea Woods
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Karla Parra
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Zoi E Sychev
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Alice Mazzagatti
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Xiaofang Huo
- Eugene McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, USA
| | - Lan Yu
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Collin Gilbreath
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Wei-Min Chen
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Anthony J Davis
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Peter Ly
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Justin M Drake
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Ralf Kittler
- Eugene McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, USA.
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA.
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
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
|
14
|
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
|
15
|
Mao N, Lee YS, Salsabeel N, Zhang Z, Li D, Kaur H, Chen X, Chang Q, Mehta S, Barnes J, de Stanchina E, Garippa R, Chen Y, Sawyers C, Carver BS. Uncoupling of Akt and mTOR signaling drives resistance to Akt inhibition in PTEN loss prostate cancers. SCIENCE ADVANCES 2025; 11:eadq3802. [PMID: 39919177 PMCID: PMC11804928 DOI: 10.1126/sciadv.adq3802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 01/09/2025] [Indexed: 02/09/2025]
Abstract
Recent phase 3 clinical trial showed improved radiographic progression-free survival in PTEN-deficient prostate cancers treated with combined Akt and AR inhibition. Building on this and our previous research into PI3K and AR signaling interactions, we aimed to define the mechanisms of response and resistance to Akt inhibition. We discovered that restoration of mTOR signaling was the early dominant driver of resistance to Akt inhibition. Mechanistically, this can be achieved through molecular alterations, resulting in loss of negative regulators of mTOR. Unexpectedly, we discovered that this was dominated by restoration of mTOR signaling through the nutrient sensing arm. This can be achieved by loss of the components of the GATOR/KICSTOR complexes or through cellular processes, leading to the recycling of amino acids. The addition of an mTOR inhibitor restored sensitivity to Akt inhibition and represents a precision-based strategy to overcome resistance in the clinic.
Collapse
Affiliation(s)
- Ninghui Mao
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Young Sun Lee
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Nazifa Salsabeel
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Zeda Zhang
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Dan Li
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Harmanpreet Kaur
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Xiaoping Chen
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Qing Chang
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sanjay Mehta
- The Gene Editing and Screening Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jesse Barnes
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ralph Garippa
- The Gene Editing and Screening Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yu Chen
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Charles Sawyers
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Howard Hughes Medical Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Brett S. Carver
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Division of Urology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
16
|
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
|
17
|
de Bono JS, He M, Shi Z, Nowicka M, Bracarda S, Sternberg CN, Chi KN, Olmos D, Sandhu S, Massard C, Matsubara N, Chen G, Bienz NS, Canter D, Wongchenko M, Sweeney C. Final Overall Survival and Molecular Data Associated with Clinical Outcomes in Patients Receiving Ipatasertib and Abiraterone in the Phase 3 IPATential150 Trial. Eur Urol 2025:S0302-2838(24)02771-4. [PMID: 39884884 DOI: 10.1016/j.eururo.2024.12.015] [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: 07/22/2024] [Revised: 10/29/2024] [Accepted: 12/19/2024] [Indexed: 02/01/2025]
Abstract
BACKGROUND AND OBJECTIVE In the phase 3 IPATential150 trial, ipatasertib addition to abiraterone significantly reduced the risk of disease progression in men with metastatic castration-resistant prostate cancer (mCRPC) with PTEN loss on immunohistochemistry (IHC), but not in the intention-to-treat (ITT) population. Here we report the final overall survival (OS) analysis and present results for prespecified and exploratory biomarker analyses. METHODS Patients were randomized to receive ipatasertib (400 mg once daily) or placebo. All patients received abiraterone (1000 mg once daily) and prednisone (5 mg twice daily). OS was assessed in patients with PTEN loss on IHC and the ITT population. Exploratory biomarker analyses included PTEN status via next-generation sequencing (NGS) and other key genomic alterations. KEY FINDINGS AND LIMITATIONS At final analysis (median follow-up 33.9 mo), ipatasertib addition did not improve OS for patients with PTEN loss in IHC (n = 521; stratified hazard ratio [sHR] 0.94, 95% confidence interval [CI] 0.76-1.17; p = 0.57) or the ITT population (n = 1101; sHR 0.91, 95% CI 0.79-1.07; not formally tested). Exploratory NGS assessments identified subgroups with genomic PTEN loss (n = 208) or PIK3CA/AKT1/PTEN alterations (n = 250), with potentially better outcomes from ipatasertib (HR 0.76, 95% CI 0.54-1.07; and HR 0.70, 95% CI 0.51-0.96, respectively). Limitations include the exploratory nature of the analysis, incomplete availability of NGS data, and potential intrapatient heterogeneity. CONCLUSIONS AND CLINICAL IMPLICATIONS Ipatasertib addition to abiraterone did not improve OS for men with mCRPC, regardless of PTEN status on IHC. Exploratory biomarker analyses identified additional genomic alterations of potential clinical relevance for AKT blockade in mCRPC that require further validation in prospective studies.
Collapse
Affiliation(s)
- Johann S de Bono
- Institute of Cancer Research and Royal Marsden Hospital London UK
| | - Meng He
- Genentech South San Francisco CA USA
| | - Zhen Shi
- Genentech South San Francisco CA USA
| | | | | | - Cora N Sternberg
- Englander Institute for Precision Medicine, Weill Cornell Medicine, Meyer Cancer Center, NewYork-Presbyterian New York NY USA
| | | | - David Olmos
- Instituto de Investigación Sanitaria, Hospital Universitario 12 de Octubre Madrid Spain
| | - Shahneen Sandhu
- Peter MacCallum Cancer Centre and University of Melbourne Melbourne Australia
| | | | | | - Geng Chen
- Genentech South San Francisco CA USA
| | | | | | | | - Christopher Sweeney
- South Australian Immunogenomics Cancer Institute, University of Adelaide Adelaide Australia.
| |
Collapse
|
18
|
Muniyan S, Vengoji R, Nimmakayala RK, Seshacharyulu P, Perumalsamy B, Alsafwani ZW, Kakar SS, Smith LM, Shonka N, Teply BA, Lele SM, Ponnusamy MP, Batra SK. PAF1-mediated transcriptional reprogramming confers docetaxel resistance in advanced prostate cancer. Cancer Lett 2025; 609:217355. [PMID: 39603380 PMCID: PMC11912633 DOI: 10.1016/j.canlet.2024.217355] [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: 03/21/2024] [Revised: 11/08/2024] [Accepted: 11/24/2024] [Indexed: 11/29/2024]
Abstract
Advanced prostate cancer (PCa) remains a significant clinical challenge, and docetaxel plays a significant role in disease management. Despite the efficacy of docetaxel as a first-line chemotherapy, resistance often develops. We developed three clinically relevant in vitro PCa cell models and transcriptomic analysis identified that the Paf1/RNA polymerase II complex component (PAF1)-associated pluripotent-transcription factor (TF), SOX2, plays a crucial role in docetaxel resistance. The cancer stem cell (CSC) transcriptional master regulator PAF1 is significantly higher in PCa cell lines, tumor tissues, and docetaxel resistant (DR) PCa cells than in age-matched control cells. To determine the molecular underlying and functional characteristics of PAF1 in resistance mechanisms, we performed coimmunoprecipitation, embryonic stem cell network proteins, in vitro tumor-initiating ability, and 3D multicellular organoid growth using PAF1 knockdown cells. Tet-inducible PAF1 depletion reduced the drug-efflux phenotype, tumor-initiating frequencies, and three-dimensional organoid growth of the docetaxel-resistant PCa cell lines. Functional studies also showed restoration of docetaxel sensitivity in a 3D tumorsphere model upon PAF1 depletion. PAF1 depletion was also associated with decreased pluripotent TFs and other CSC markers. This study provides a novel regulatory mechanism of docetaxel resistance in PCa through PAF1.
Collapse
Affiliation(s)
- Sakthivel Muniyan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Raghupathy Vengoji
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Rama Krishna Nimmakayala
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Parthasarathy Seshacharyulu
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Balaji Perumalsamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Zahraa Wajih Alsafwani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Sham S Kakar
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Lynette M Smith
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Nicole Shonka
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Department of Internal Medicine, Division of Oncology and Hematology, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Benjamin A Teply
- Department of Internal Medicine, Division of Oncology and Hematology, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Subodh M Lele
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
19
|
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
|
20
|
Constâncio V, Lobo J, Sequeira JP, Henrique R, Jerónimo C. Prostate cancer epigenetics - from pathophysiology to clinical application. Nat Rev Urol 2025:10.1038/s41585-024-00991-8. [PMID: 39820138 DOI: 10.1038/s41585-024-00991-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2024] [Indexed: 01/19/2025]
Abstract
Prostate cancer is a multifactorial disease influenced by various molecular features. Over the past decades, epigenetics, which is the study of changes in gene expression without altering the DNA sequence, has been recognized as a major driver of this disease. In the past 50 years, advancements in technological tools to characterize the epigenome have highlighted crucial roles of epigenetic mechanisms throughout the entire spectrum of prostate cancer, from initiation to progression, including localized disease, metastatic dissemination, castration resistance and neuroendocrine transdifferentiation. Substantial advances in the understanding of epigenetic mechanisms in the pathophysiology of prostate cancer have been carried out, but translating preclinical achievements into clinical practice remains challenging. Ongoing research and biomarker-oriented clinical trials are expected to increase the likelihood of successfully integrating epigenetics into prostate cancer clinical management.
Collapse
Affiliation(s)
- Vera Constâncio
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), Porto, Portugal
- Doctoral Program in Biomedical Sciences, ICBAS - School of Medicine & Biomedical Sciences, University of Porto (ICBAS-UP), Porto, Portugal
| | - João Lobo
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), Porto, Portugal
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre Raquel Seruca (Porto.CCC), Porto, Portugal
- Department of Pathology and Molecular Immunology, ICBAS - School of Medicine & Biomedical Sciences, University of Porto, Porto, Portugal
| | - José Pedro Sequeira
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), Porto, Portugal
- Doctoral Program in Biomedical Sciences, ICBAS - School of Medicine & Biomedical Sciences, University of Porto (ICBAS-UP), Porto, Portugal
| | - Rui Henrique
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), Porto, Portugal
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre Raquel Seruca (Porto.CCC), Porto, Portugal
- Department of Pathology and Molecular Immunology, ICBAS - School of Medicine & Biomedical Sciences, University of Porto, Porto, Portugal
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), Porto, Portugal.
- Department of Pathology and Molecular Immunology, ICBAS - School of Medicine & Biomedical Sciences, University of Porto, Porto, Portugal.
| |
Collapse
|
21
|
Chauhan PS, Alahi I, Sinha S, Ledet EM, Mueller R, Linford J, Shiang AL, Webster J, Greiner L, Yang B, Ni G, Dang HX, Saha D, Babbra RK, Feng W, Harris PK, Qaium F, Duose DY, Alexander SE, Sherry AD, Jaeger EB, Miller PJ, Caputo SA, Orme JJ, Lucien F, Park SS, Tang C, Pachynski RK, Sartor O, Maher CA, Chaudhuri AA. Genomic and Epigenomic Analysis of Plasma Cell-Free DNA Identifies Stemness Features Associated with Worse Survival in Lethal Prostate Cancer. Clin Cancer Res 2025; 31:151-163. [PMID: 39177583 PMCID: PMC11743868 DOI: 10.1158/1078-0432.ccr-24-1658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/21/2024] [Accepted: 08/21/2024] [Indexed: 08/24/2024]
Abstract
PURPOSE Metastatic castration-resistant prostate cancer (mCRPC) resistant to androgen receptor signaling inhibitors (ARSI) is often lethal. Liquid biopsy biomarkers for this deadly form of disease remain under investigation, and underpinning mechanisms remain ill-understood. EXPERIMENTAL DESIGN We applied targeted cell-free DNA (cfDNA) sequencing to 126 patients with mCRPC from three academic cancer centers and separately performed genome-wide cfDNA methylation sequencing on 43 plasma samples collected prior to the initiation of first-line ARSI treatment. To analyze the genome-wide sequencing data, we performed nucleosome positioning and differential methylated region analysis. We additionally analyzed single-cell and bulk RNA sequencing data from 14 and 80 patients with mCRPC, respectively, to develop and validate a stem-like signature, which we inferred from cfDNA. RESULTS Targeted cfDNA sequencing detected AR/enhancer alterations prior to first-line ARSIs that correlated with significantly worse progression-free survival (P = 0.01; HR = 2.12) and overall survival (P = 0.02; HR = 2.48). Plasma methylome analysis revealed that AR/enhancer lethal mCRPC patients have significantly higher promoter-level hypomethylation than AR/enhancer wild-type mCRPC patients (P < 0.0001). Moreover, gene ontology and CytoTRACE analysis of nucleosomally more accessible transcription factors in cfDNA revealed enrichment for stemness-associated transcription factors in patients with lethal mCRPC. The resulting stemness signature was then validated in a completely held-out cohort of 80 patients with mCRPC profiled by tumor RNA sequencing. CONCLUSIONS We analyzed a total of 220 patients with mCRPC, validated the importance of cell-free AR/enhancer alterations as a prognostic biomarker in lethal mCRPC, and showed that the underlying mechanism for lethality involves reprogramming developmental states toward increased stemness. See related commentary by Nawfal et al., p. 7.
Collapse
Affiliation(s)
- Pradeep S. Chauhan
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, United States of America
| | - Irfan Alahi
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, United States of America
- Department of Computer Science and Engineering, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Savar Sinha
- Division of Cancer Biology, Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Elisa M. Ledet
- Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Ryan Mueller
- Division of Cancer Biology, Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Jessica Linford
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, United States of America
| | | | - Jace Webster
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Lilli Greiner
- Division of Cancer Biology, Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Breanna Yang
- Division of Cancer Biology, Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Gabris Ni
- Division of Cancer Biology, Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Ha X. Dang
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
- McDonnell Genome Institute, Washington University in St. Louis, Missouri, United States of America
| | - Debanjan Saha
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Ramandeep K. Babbra
- Wilmot Institute Cancer Center, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Wenjia Feng
- Division of Cancer Biology, Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Peter K. Harris
- Division of Cancer Biology, Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Faridi Qaium
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, United States of America
| | - Dzifa Y. Duose
- Department of Radiation Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Sanchez E. Alexander
- Department of Radiation Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Alexander D. Sherry
- Department of Radiation Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Ellen B. Jaeger
- Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Patrick J. Miller
- Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Sydney A. Caputo
- Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Jacob J. Orme
- Division of Oncology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
- Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota, United States of America
| | - Fabrice Lucien
- Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota, United States of America
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Urology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Sean S. Park
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, United States of America
- Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota, United States of America
| | - Chad Tang
- Department of Radiation Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Russell K. Pachynski
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Siteman Cancer Center, Washington University in St. Louis, Missouri, United States of America
| | - Oliver Sartor
- Division of Oncology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Urology, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Christopher A. Maher
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
- McDonnell Genome Institute, Washington University in St. Louis, Missouri, United States of America
- Siteman Cancer Center, Washington University in St. Louis, Missouri, United States of America
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Aadel A. Chaudhuri
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, United States of America
- Department of Computer Science and Engineering, Washington University in St. Louis, St. Louis, Missouri, United States of America
- Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota, United States of America
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
| |
Collapse
|
22
|
Liu K, Li Y, Shen M, Xu W, Wu S, Yang X, Zhang B, Lin N. Epigenetic Regulation of Stromal and Immune Cells and Therapeutic Targets in the Tumor Microenvironment. Biomolecules 2025; 15:71. [PMID: 39858465 PMCID: PMC11764280 DOI: 10.3390/biom15010071] [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: 11/18/2024] [Revised: 12/19/2024] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
The tumor microenvironment (TME) plays a pivotal role in neoplastic initiation and progression. Epigenetic machinery, governing the expression of core oncogenes and tumor suppressor genes in transformed cells, significantly contributes to tumor development at both primary and distant sites. Recent studies have illuminated how epigenetic mechanisms integrate external cues and downstream signals, altering the phenotype of stromal cells and immune cells. This remolds the area surrounding tumor cells, ultimately fostering an immunosuppressive microenvironment. Therefore, correcting the TME by targeting the epigenetic modifications holds substantial promise for cancer treatment. This review synthesizes recent research that elucidates the impact of specific epigenetic regulations-ranging from DNA methylation to histone modifications and chromatin remodeling-on stromal and immune cells within the TME. Notably, we highlight their functional roles in either promoting or restricting tumor progression. We also discuss the potential applications of epigenetic agents for cancer treatment, envisaging their ability to normalize the ecosystem. This review aims to assist researchers in understanding the dynamic interplay between epigenetics and the TME, paving the way for better epigenetic therapy.
Collapse
Affiliation(s)
- Kang Liu
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Yue Li
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Minmin Shen
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Drug Clinical Trial Institution, Huzhou Central Hospital, Huzhou 313000, China
| | - Wei Xu
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Shanshan Wu
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Xinxin Yang
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Bo Zhang
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Nengming Lin
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
- Westlake Laboratory of Life Sciences and Biomedicine of Zhejiang Province, Westlake University, Hangzhou 310024, China
| |
Collapse
|
23
|
Li F, Dai P, Shi H, Zhang Y, He J, Gopalan A, Li D, Chen Y, Du Y, Xu G, Yang W, Liang C, Gao D. LKB1 inactivation promotes epigenetic remodeling-induced lineage plasticity and antiandrogen resistance in prostate cancer. Cell Res 2025; 35:59-71. [PMID: 39743630 PMCID: PMC11701123 DOI: 10.1038/s41422-024-01025-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 08/22/2024] [Indexed: 01/04/2025] Open
Abstract
Epigenetic regulation profoundly influences the fate of cancer cells and their capacity to switch between lineages by modulating essential gene expression, thereby shaping tumor heterogeneity and therapy response. In castration-resistant prostate cancer (CRPC), the intricacies behind androgen receptor (AR)-independent lineage plasticity remain unclear, leading to a scarcity of effective clinical treatments. Utilizing single-cell RNA sequencing on both human and mouse prostate cancer samples, combined with whole-genome bisulfite sequencing and multiple genetically engineered mouse models, we investigated the molecular mechanism of AR-independent lineage plasticity and uncovered a potential therapeutic strategy. Single-cell transcriptomic profiling of human prostate cancers, both pre- and post-androgen deprivation therapy, revealed an association between liver kinase B1 (LKB1) pathway inactivation and AR independence. LKB1 inactivation led to AR-independent lineage plasticity and global DNA hypomethylation during prostate cancer progression. Importantly, the pharmacological inhibition of TET enzymes and supplementation with S-adenosyl methionine were found to effectively suppress AR-independent prostate cancer growth. These insights shed light on the mechanism driving AR-independent lineage plasticity and propose a potential therapeutic strategy by targeting DNA hypomethylation in AR-independent CRPC.
Collapse
MESH Headings
- Male
- Animals
- Humans
- Epigenesis, Genetic/drug effects
- Mice
- Androgen Antagonists/pharmacology
- Androgen Antagonists/therapeutic use
- Protein Serine-Threonine Kinases/metabolism
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/drug effects
- Receptors, Androgen/metabolism
- Receptors, Androgen/genetics
- AMP-Activated Protein Kinase Kinases
- DNA Methylation/drug effects
- Cell Line, Tumor
- Prostatic Neoplasms, Castration-Resistant/pathology
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/metabolism
- Prostatic Neoplasms/pathology
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/drug therapy
- Prostatic Neoplasms/metabolism
- Cell Lineage
- Gene Expression Regulation, Neoplastic/drug effects
- Cell Plasticity/drug effects
- AMP-Activated Protein Kinases
Collapse
Affiliation(s)
- Fei Li
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Pengfei Dai
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Huili Shi
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yajuan Zhang
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juan He
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Anuradha Gopalan
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dan Li
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yu Chen
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yarui Du
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Guoliang Xu
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Weiwei Yang
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
| | - Chao Liang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Dong Gao
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China.
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China.
| |
Collapse
|
24
|
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
|
25
|
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
|
26
|
Deng B, Ke B, Tian Q, Gao Y, Zhai Q, Zhang W. Targeting AURKA with multifunctional nanoparticles in CRPC therapy. J Nanobiotechnology 2024; 22:803. [PMID: 39734237 DOI: 10.1186/s12951-024-03070-7] [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: 08/04/2024] [Accepted: 12/11/2024] [Indexed: 12/31/2024] Open
Abstract
Castration-resistant prostate cancer (CRPC) presents significant therapeutic challenges due to its aggressive nature and poor prognosis. Targeting Aurora-A kinase (AURKA) has shown promise in cancer treatment. This study investigates the efficacy of ART-T cell membrane-encapsulated AMS@AD (CM-AMS@AD) nanoparticles (NPs) in a photothermal-chemotherapy-immunotherapy combination for CRPC. Bioinformatics analysis of the Cancer Genome Atlas-prostate adenocarcinoma (TCGA-PRAD) dataset revealed overexpression of AURKA in PCa, correlating with poor clinical outcomes. Single-cell RNA sequencing data from the GEO database showed a significant reduction in immune cells in CRPC. Experimentally, T cell membrane-biomimetic NPs loaded with the AURKA inhibitor Alisertib and chemotherapy drug DTX were synthesized and characterized by dynamic light scattering and transmission electron microscopy, showing good stability and uniformity (average diameter: 158 nm). In vitro studies demonstrated that these NPs inhibited CRPC cell proliferation, increased the G2/M cell population, and elevated apoptosis, confirmed by γH2AX expression. In vivo, CM-AMS@AD NPs accumulated in tumor tissues, significantly slowed tumor growth, decreased proliferation, increased apoptosis, and improved the immune environment, enhancing dendritic cell (DC) maturation and increasing CD8 + /CD4 + ratios. These findings suggest that CM-AMS@AD NPs offer a promising triple-combination therapy for CRPC, integrating photothermal, chemotherapy, and immunotherapy, with significant potential for future clinical applications.
Collapse
Affiliation(s)
- Bin Deng
- Department of Urology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, China
- Department of Science and Technology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, China
| | - Binghu Ke
- Department of Urology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, China
| | - Qixing Tian
- Department of Urology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, China
| | - Yukui Gao
- Department of Urology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, China.
| | - Qiliang Zhai
- Department of Urology, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, Jiangxi, China.
| | - Wenqiang Zhang
- Department of Urology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, China.
| |
Collapse
|
27
|
Teng J, Jia Z, Gao F, Guan Y, Yao L, Ma C, Li Z, Ai X. AP-1 activates KCNN4-mediated Ca2 + signaling to induce the Th1/Th17 differentiation of CD4 + cells in chronic non-bacterial prostatitis. Cell Biol Toxicol 2024; 41:18. [PMID: 39729199 PMCID: PMC11680619 DOI: 10.1007/s10565-024-09967-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024]
Abstract
The intraprostatic inflammatory infiltrate is characterized by Th1 CD4+ T cells, and its molecular mechanism is not well defined. This study explored the mechanisms responsible for the alteration of Th1/Th17 differentiation of CD4+ T cells in chronic non-bacterial prostatitis (CNP). CNP rats were induced by the administration of testosterone and 17β-estradiol. The Th1/Th17 cell percentage was increased in the prostate tissue of CNP rats, which was accompanied by increased IL-2, IFN-γ, IL-17A, and IL-22 levels. Transcriptome sequencing was performed, followed by KEGG pathway enrichment analysis. Activator protein-1 (AP-1) was enhanced in CD4+ T cells from CNP rats, and its inhibitor SR11302 suppressed Th1/Th17 differentiation and delayed CNP. AP-1 transcriptionally activated the expression of KCNN4, which potentiated mTORC1 in CD4+ T cells by enhancing Ca2+ signaling, thereby promoting Th1/Th17 differentiation. Rapamycin-mediated autophagy activation reversed AP-1/KCNN4/mTORC1-promoted Th1/Th17 differentiation, thereby inhibiting CNP. These results suggest that AP-1-mediated KCNN4 transcription promotes the inhibition of autophagy by mTORC1 through Ca2+ signaling, which supports Th1/Th17 differentiation of CD4+ T cells, resulting in the transformation of CNP to prostatic intraepithelial neoplasia and adenocarcinoma.
Collapse
Affiliation(s)
- Jingfei Teng
- Department of Urology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, P.R. China
- Department of Urology, The Third Medical Center of Chinese PLA General Hospital, Beijing, 100039, P.R. China
| | - Zhuomin Jia
- Department of Urology, The Third Medical Center of Chinese PLA General Hospital, Beijing, 100039, P.R. China
| | - Feng Gao
- Department of Urology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, P.R. China
| | - Yawei Guan
- Department of Urology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, P.R. China
- Department of Urology, The Third Medical Center of Chinese PLA General Hospital, Beijing, 100039, P.R. China
| | - Li Yao
- Department of Urology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, P.R. China
| | - Chong Ma
- Department of Urology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, P.R. China
- Department of Urology, The Third Medical Center of Chinese PLA General Hospital, Beijing, 100039, P.R. China
| | - Zhihui Li
- Department of Urology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, P.R. China
| | - Xing Ai
- Department of Urology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, P.R. China.
- Department of Urology, The Third Medical Center of Chinese PLA General Hospital, Beijing, 100039, P.R. China.
| |
Collapse
|
28
|
Cho H, Zhang Y, Tien JC, Mannan R, Luo J, Narayanan SP, Mahapatra S, Hu J, Shelley G, Cruz G, Shahine M, Wang L, Su F, Wang R, Cao X, Dhanasekaran SM, Keller ET, Pitchiaya S, Chinnaiyan AM. Cellular cartography reveals mouse prostate organization and determinants of castration resistance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.27.630532. [PMID: 39763898 PMCID: PMC11703157 DOI: 10.1101/2024.12.27.630532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Inadequate response to androgen deprivation therapy (ADT) frequently arises in prostate cancer, driven by cellular mechanisms that remain poorly understood. Here, we integrated single-cell RNA sequencing, single-cell multiomics, and spatial transcriptomics to define the transcriptional, epigenetic, and spatial basis of cell identity and castration response in the mouse prostate. Leveraging these data along with a meta-analysis of human prostates and prostate cancer, we identified cellular orthologs and key determinants of ADT response and resistance. Our findings reveal that mouse prostates harbor lobe-specific luminal epithelial cell types distinguished by unique gene regulatory modules and anatomically defined androgen-responsive transcriptional programs, indicative of divergent developmental origins. Androgen-insensitive, stem-like epithelial populations - resembling human club and hillock cells - are notably enriched in the urethra and ventral prostate but are rare in other lobes. Within the ventral prostate, we also uncovered two additional androgen-responsive luminal epithelial cell types, marked by Pbsn or Spink1 expression, which align with human luminal subsets and may define the origin of distinct prostate cancer subtypes. Castration profoundly reshaped luminal epithelial transcriptomes, with castration-resistant luminal epithelial cells activating stress-responsive and stemness programs. These transcriptional signatures are enriched in tumor cells from ADT-treated and castration-resistant prostate cancer patients, underscoring their likely role in driving treatment resistance. Collectively, our comprehensive cellular atlas of the mouse prostate illuminates the importance of lobe-specific contexts for prostate cancer modeling and reveals potential therapeutic targets to counter castration resistance.
Collapse
Affiliation(s)
- Hanbyul Cho
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, 48109
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109
| | - Yuping Zhang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, 48109
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109
| | - Jean C. Tien
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, 48109
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109
| | - Rahul Mannan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, 48109
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109
| | - Jie Luo
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, 48109
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109
| | - Sathiya Pandi Narayanan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, 48109
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109
| | - Somnath Mahapatra
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, 48109
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109
| | - Jing Hu
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, 48109
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109
- Department of Pathology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Greg Shelley
- Department of Urology, University of Michigan, Ann Arbor, MI, 48109
| | - Gabriel Cruz
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, 48109
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109
| | - Miriam Shahine
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, 48109
| | - Lisha Wang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, 48109
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109
| | - Fengyun Su
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, 48109
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109
| | - Rui Wang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, 48109
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109
| | - Xuhong Cao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, 48109
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109
| | - Saravana Mohan Dhanasekaran
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, 48109
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109
- Department of Urology, University of Michigan, Ann Arbor, MI, 48109
| | - Evan T. Keller
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109
- Department of Urology, University of Michigan, Ann Arbor, MI, 48109
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109
- Single Cell Spatial Analysis Program, University of Michigan, Ann Arbor, MI, 48109
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109
| | - Sethuramasundaram Pitchiaya
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, 48109
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109
- Department of Pathology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Urology, University of Michigan, Ann Arbor, MI, 48109
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109
| | - Arul M. Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, 48109
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109
- Department of Urology, University of Michigan, Ann Arbor, MI, 48109
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, 48109
| |
Collapse
|
29
|
Senatorov IS, Bowman J, Jansson KH, Alilin AN, Capaldo BJ, Lake R, Riba M, Abbey YC, Mcknight C, Zhang X, Raj S, Beshiri ML, Shinn P, Nguyen H, Thomas CJ, Corey E, Kelly K. Castrate-resistant prostate cancer response to taxane is determined by an HNF1-dependent apoptosis resistance circuit. Cell Rep Med 2024; 5:101868. [PMID: 39657662 PMCID: PMC11722106 DOI: 10.1016/j.xcrm.2024.101868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 09/09/2024] [Accepted: 11/14/2024] [Indexed: 12/12/2024]
Abstract
Metastatic castrate-resistant prostate cancer (mCRPC) is a genetically and phenotypically heterogeneous cancer where advancements are needed in biomarker discovery and targeted therapy. A critical and often effective component of treatment includes taxanes. We perform a high-throughput screen across a cohort of 30 diverse patient-derived castrate-resistant prostate cancer (CRPC) organoids to a library of 78 drugs. Combining quantitative response measures with transcriptomic analyses demonstrates that HNF1 homeobox A (HNF1A) drives a transcriptional program of taxane resistance, commonly dependent upon cellular inhibitor of apoptosis protein 2 (cIAP2). Monotherapy with cIAP2 inhibitor LCL161 is sufficient to treat HNF1A+ models of mCRPC previously resistant to docetaxel. These data may be useful in future clinical trial designs.
Collapse
Affiliation(s)
- Ilya S Senatorov
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Joel Bowman
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Keith H Jansson
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Aian Neil Alilin
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Brian J Capaldo
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Ross Lake
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Morgan Riba
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Yasmine C Abbey
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Crystal Mcknight
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Xiaohu Zhang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Sonam Raj
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Michael L Beshiri
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Paul Shinn
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Holly Nguyen
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Craig J Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA; Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, NIH, Bethesda, MD, USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Kathleen Kelly
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA.
| |
Collapse
|
30
|
Qin H, Zhang Q, Guo Y. Genome-wide identification of alternative splicing related with transcription factors and splicing regulators in breast cancer stem cells responding to fasting-mimicking diet. Comput Biol Chem 2024; 113:108272. [PMID: 39509796 DOI: 10.1016/j.compbiolchem.2024.108272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 10/21/2024] [Accepted: 10/28/2024] [Indexed: 11/15/2024]
Abstract
Fasting-mimicking diet (FMD) can effectively inhibit the viability of breast cancer stem cells (CSCs). However, the molecular mechanisms underlying the inhibitory function of FMD on breast CSCs remain largely unknown. Elucidating the mechanisms by which FMD suppresses breast CSCs is beneficial to targeting breast CSCs. Herein, we systematically analyze alternative splicing and RNA binding protein (RBP) expression in breast CSCs during FMD. The analysis results show that a large number of regulated alternative splicing (RAS) and differentially expressed genes (DEGs) appear responding to FMD. Further studies show that there are potential regulatory relationships between transcription factors (TFs) with RAS (RAS-TFs) and their differentially expressed target genes (RAS-TF-DEGs). Moreover, differentially expressed RNA binding proteins (DERBPs) exhibit potential regulatory functions on RAS-TFs. In short, DERBPs potentially control the alternative splicing of TFs (RAS-TFs), regulating their target gene (RAS-TF-DEG) expression, which leads to the regulation of biological processes in breast CSCs during FMD. In addition, the alternative splicing and DEGs are compared between breast CSCs and differentiated cancer cells during FMD, providing new interpretations for the different responses of the two types of cells. Our studies will shed light on the understanding of the molecular mechanisms underlying breast CSC inhibition induced by FMD.
Collapse
Affiliation(s)
- Hongshuang Qin
- Department of Biological and Food Engineering, Lyuliang University, Lvliang, Shanxi 033001, China.
| | - Qian Zhang
- Department of Biological and Food Engineering, Lyuliang University, Lvliang, Shanxi 033001, China
| | - Yanxiang Guo
- Department of Biological and Food Engineering, Lyuliang University, Lvliang, Shanxi 033001, China
| |
Collapse
|
31
|
Lee S, Lee DY, So I, Chun JN, Jeon JH. Chromatin accessibility is associated with therapeutic response in prostate cancer. Oncol Lett 2024; 28:605. [PMID: 39483964 PMCID: PMC11525612 DOI: 10.3892/ol.2024.14738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 09/12/2024] [Indexed: 11/03/2024] Open
Abstract
Treatment of advanced prostate cancer is challenging due to a lack of effective therapies. Therefore, it is important to understand the molecular mechanisms underlying therapeutic resistance in prostate cancer and to identify promising drug targets offering significant clinical advantages. Given the pivotal role of dysregulated transcriptional programs in the therapeutic response, it is essential to prioritize translational efforts targeting cancer-associated transcription factors (TFs). The present study investigated whether chromatin accessibility was associated with therapeutic resistance in prostate cancer using Assay for Transposase-Accessible Chromatin with sequencing (ATAC-seq) data. The bioinformatics analysis identified differences in chromatin accessibility between the drug response (Remission) and drug resistance (Disease) groups. Additionally, a significant association was observed between chromatin accessibility, transcriptional output and TF activity. Among TFs, forkhead box protein M1 (FOXM1) was identified as a TF with high activity and expression in the Disease group. Notably, the results of the computational analysis were validated by FOXM1 knockdown experiments, which resulted in suppressed cell proliferation and enhanced therapeutic sensitivity in prostate cancer cells. The present findings demonstrated that chromatin accessibility and TF activity may be associated with therapeutic resistance in prostate cancer. Additionally, these results provide the basis for future investigations aimed at understanding the molecular mechanisms of drug resistance and developing novel therapeutic approaches for prostate cancer.
Collapse
Affiliation(s)
- Sanghoon Lee
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Da Young Lee
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Insuk So
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Republic of Korea
| | - Jung Nyeo Chun
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Republic of Korea
| | - Ju-Hong Jeon
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Republic of Korea
| |
Collapse
|
32
|
Kiviaho A, Eerola SK, Kallio HML, Andersen MK, Hoikka M, Tiihonen AM, Salonen I, Spotbeen X, Giesen A, Parker CTA, Taavitsainen S, Hantula O, Marttinen M, Hermelo I, Ismail M, Midtbust E, Wess M, Devlies W, Sharma A, Krossa S, Häkkinen T, Afyounian E, Vandereyken K, Kint S, Kesseli J, Tolonen T, Tammela TLJ, Viset T, Størkersen Ø, Giskeødegård GF, Rye MB, Murtola T, Erickson A, Latonen L, Bova GS, Mills IG, Joniau S, Swinnen JV, Voet T, Mirtti T, Attard G, Claessens F, Visakorpi T, Rautajoki KJ, Tessem MB, Urbanucci A, Nykter M. Single cell and spatial transcriptomics highlight the interaction of club-like cells with immunosuppressive myeloid cells in prostate cancer. Nat Commun 2024; 15:9949. [PMID: 39550375 PMCID: PMC11569175 DOI: 10.1038/s41467-024-54364-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 11/08/2024] [Indexed: 11/18/2024] Open
Abstract
Prostate cancer treatment resistance is a significant challenge facing the field. Genomic and transcriptomic profiling have partially elucidated the mechanisms through which cancer cells escape treatment, but their relation toward the tumor microenvironment (TME) remains elusive. Here we present a comprehensive transcriptomic landscape of the prostate TME at multiple points in the standard treatment timeline employing single-cell RNA-sequencing and spatial transcriptomics data from 120 patients. We identify club-like cells as a key epithelial cell subtype that acts as an interface between the prostate and the immune system. Tissue areas enriched with club-like cells have depleted androgen signaling and upregulated expression of luminal progenitor cell markers. Club-like cells display a senescence-associated secretory phenotype and their presence is linked to increased polymorphonuclear myeloid-derived suppressor cell (PMN-MDSC) activity. Our results indicate that club-like cells are associated with myeloid inflammation previously linked to androgen deprivation therapy resistance, providing a rationale for their therapeutic targeting.
Collapse
Affiliation(s)
- Antti Kiviaho
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - Sini K Eerola
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - Heini M L Kallio
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - Maria K Andersen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Miina Hoikka
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - Aliisa M Tiihonen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - Iida Salonen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - Xander Spotbeen
- Laboratory of Lipid Metabolism and Cancer, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
- KU Leuven Institute for Single Cell Omics (LISCO), KU Leuven, Leuven, Belgium
| | - Alexander Giesen
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | | | - Sinja Taavitsainen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - Olli Hantula
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - Mikael Marttinen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, Heidelberg, Germany
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| | - Ismaïl Hermelo
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | | | - Elise Midtbust
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Maximilian Wess
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Wout Devlies
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
- Molecular Endocrinology Laboratory, Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Abhibhav Sharma
- Department of Public Health and Nursing, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Sebastian Krossa
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- Central staff, St. Olavs Hospital HF, 7006, Trondheim, Norway
| | - Tomi Häkkinen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - Ebrahim Afyounian
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - Katy Vandereyken
- KU Leuven Institute for Single Cell Omics (LISCO), KU Leuven, Leuven, Belgium
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Sam Kint
- KU Leuven Institute for Single Cell Omics (LISCO), KU Leuven, Leuven, Belgium
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Juha Kesseli
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - Teemu Tolonen
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
- Department of Pathology, Fimlab Laboratories, Ltd, Tampere University Hospital, Tampere, Finland
| | - Teuvo L J Tammela
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Urology, Tampere University Hospital, Tampere, Finland
| | - Trond Viset
- Department of Pathology, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Øystein Størkersen
- Department of Pathology, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Guro F Giskeødegård
- Clinic of Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Public Health and Nursing, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Morten B Rye
- Clinic of Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Teemu Murtola
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - Andrew Erickson
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- ICAN-Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Leena Latonen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - G Steven Bova
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - Ian G Mills
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
- Patrick G Johnston Centre for Cancer Research, Queen's University of Belfast, Belfast, UK
| | - Steven Joniau
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Johannes V Swinnen
- Laboratory of Lipid Metabolism and Cancer, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
- KU Leuven Institute for Single Cell Omics (LISCO), KU Leuven, Leuven, Belgium
| | - Thierry Voet
- KU Leuven Institute for Single Cell Omics (LISCO), KU Leuven, Leuven, Belgium
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Tuomas Mirtti
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- ICAN-Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Department of Pathology, University of Helsinki & Helsinki University Hospital, Helsinki, Finland
| | - Gerhardt Attard
- University College London Cancer Institute, London, UK
- University College London Hospitals, London, UK
| | - Frank Claessens
- Molecular Endocrinology Laboratory, Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Tapio Visakorpi
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
- Fimlab Laboratories, Ltd, Tampere University Hospital, Tampere, Finland
| | - Kirsi J Rautajoki
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland
| | - May-Britt Tessem
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Alfonso Urbanucci
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland.
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
| | - Matti Nykter
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
- Prostate Cancer Research Center, Tampere University and TAYS Cancer Center, Tampere, Finland.
| |
Collapse
|
33
|
Wang L, Kong B, Wang J, Yang G, Wu X, Zang J, Li C, Wang X, Si M, Wang Z, Liu P, Wang Y, Chen H, Liu F, Yang PP, Wang L, Wang H, Xu W. A Novel Targeted Microtubules Transformable Nanopeptide System Yields Strong Anti-Prostate Cancer Effects by Suppressing Nuclear Translocation of Androgen Receptors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407826. [PMID: 39313982 DOI: 10.1002/adma.202407826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 09/06/2024] [Indexed: 09/25/2024]
Abstract
The extended use of androgen deprivation therapy (ADT) may often lead to the progression from castration-sensitive prostate cancer (CSPC) to castration-resistant prostate cancer (CRPC) in prostate cancer. To address this, it is essential to inhibit the nuclear translocation of the androgen receptor (AR) as part of an effective disease-modifying strategy. Microtubules play a central role in facilitating AR nuclear translocation, highlighting their importance as a therapeutic target. In this regard, a designated as the targeted microtubules transformable nanopeptide system (MTN) is developed. This system is designed to disrupt microtubule structure and function through dual-targeting of prostate-specific membrane antigen (PSMA) and β-tubulin. Initially, MTN targets prostate cells via PSMA and then specifically binds to β-tubulin within microtubules, leading to the formation of nanofibers. These nanofibers subsequently induce the polymerization of microtubules, thereby disrupting AR transport. Notably, MTN exhibits efficient and prolonged suppression of prostate cancer across the spectrum from CSPC to CRPC, with a highly favorable safety profile in normal cells. These findings highlight the potential of MTN as a novel and promising approach for comprehensive prostate cancer therapy throughout its entire progression.
Collapse
Affiliation(s)
- Lu Wang
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, China
| | - Bin Kong
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, China
| | - Jiaqi Wang
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, China
| | - Guang Yang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Xiuhai Wu
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, China
| | - Jiahui Zang
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, China
| | - Cong Li
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, China
| | - Xinyue Wang
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, China
| | - Minggui Si
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, China
| | - Zhijia Wang
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, China
| | - Pan Liu
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, China
| | - Yuting Wang
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, China
| | - Huilin Chen
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, China
| | - Feng Liu
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, China
| | - Pei-Pei Yang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Lei Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Wanhai Xu
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, China
- Department of Urology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| |
Collapse
|
34
|
Li Z, Li Z, Luo Y, Chen W, Fang Y, Xiong Y, Zhang Q, Yuan D, Yan B, Zhu J. Application and new findings of scRNA-seq and ST-seq in prostate cancer. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:23. [PMID: 39470950 PMCID: PMC11522250 DOI: 10.1186/s13619-024-00206-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 10/12/2024] [Indexed: 11/01/2024]
Abstract
Prostate cancer is a malignant tumor of the male urological system with the highest incidence rate in the world, which seriously threatens the life and health of middle-aged and elderly men. The progression of prostate cancer involves the interaction between tumor cells and tumor microenvironment. Understanding the mechanisms of prostate cancer pathogenesis and disease progression is important to guide diagnosis and therapy. The emergence of single-cell RNA sequencing (scRNA-seq) and spatial transcriptome sequencing (ST-seq) technologies has brought breakthroughs in the study of prostate cancer. It makes up for the defects of traditional techniques such as fluorescence-activated cell sorting that are difficult to elucidate cell-specific gene expression. This review summarized the heterogeneity and functional changes of prostate cancer and tumor microenvironment revealed by scRNA-seq and ST-seq, aims to provide a reference for the optimal diagnosis and treatment of prostate cancer.
Collapse
Affiliation(s)
- Zhuang Li
- Department of Urology, Affiliated Hospital of Guizhou Medical University, Guiyang city, 550004, Guizhou Province, China
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang city, 550002, Guizhou Province, China
| | - Zhengnan Li
- Graduate School of Zunyi Medical University, Zunyi City, 563099, Guizhou Province, China
| | - Yuanyuan Luo
- Medical College of Guizhou University, Guiyang city, 550025, Guizhou Province, China
| | - Weiming Chen
- Medical College of Guizhou University, Guiyang city, 550025, Guizhou Province, China
| | - Yinyi Fang
- Medical College of Guizhou University, Guiyang city, 550025, Guizhou Province, China
| | - Yuliang Xiong
- Department of Urology, Affiliated Hospital of Guizhou Medical University, Guiyang city, 550004, Guizhou Province, China
| | - Qinyi Zhang
- Graduate School of Zunyi Medical University, Zunyi City, 563099, Guizhou Province, China
| | - Dongbo Yuan
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang city, 550002, Guizhou Province, China
| | - Bo Yan
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang city, 550002, Guizhou Province, China
| | - Jianguo Zhu
- Department of Urology, Affiliated Hospital of Guizhou Medical University, Guiyang city, 550004, Guizhou Province, China.
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang city, 550002, Guizhou Province, China.
- Graduate School of Zunyi Medical University, Zunyi City, 563099, Guizhou Province, China.
- Medical College of Guizhou University, Guiyang city, 550025, Guizhou Province, China.
| |
Collapse
|
35
|
Cooper PO, Yang J, Wang HH, Broman MM, Jayasundara SM, Sahoo SS, Yan B, Awdalkreem GD, Cresswell GM, Wang L, Goossens E, Lanman NA, Doerge RW, Zheng F, Cheng L, Alqahtani S, Crist SA, Braun RE, Kazemian M, Jerde TJ, Ratliff TL. Inflammation impacts androgen receptor signaling in basal prostate stem cells through interleukin 1 receptor antagonist. Commun Biol 2024; 7:1390. [PMID: 39455902 PMCID: PMC11511867 DOI: 10.1038/s42003-024-07071-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: 12/04/2023] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Chronic prostate inflammation in patients with benign prostate hyperplasia (BPH) correlates with the severity of symptoms. How inflammation contributes to prostate enlargement and/or BPH symptoms and the underlying mechanisms remain unclear. In this study, we utilize a unique transgenic mouse model that mimics chronic non-bacterial prostatitis in men and investigate the impact of inflammation on androgen receptor (AR) in basal prostate stem cells (bPSC) and their differentiation in vivo. We find that inflammation significantly enhances AR levels and activity in bPSC. More importantly, we identify interleukin 1 receptor antagonist (IL-1RA) as a crucial regulator of AR in bPSC during inflammation. IL-1RA is one of the top molecules upregulated by inflammation, and inhibiting IL-1RA reverses the enhanced AR activity in organoids derived from inflamed bPSC. Additionally, IL-1RA appears to activate AR by counteracting IL-1α's inhibitory effect. Furthermore, using a lineage tracing model, we observe that inflammation induces bPSC proliferation and differentiation into luminal cells even under castrate conditions, indicating that AR activation driven by inflammation is sufficient to promote bPSC proliferation and differentiation. Taken together, our study uncovers mechanisms through which inflammation modulates AR signaling in bPSC and induces bPSC luminal differentiation that may contribute to prostate hyperplasia.
Collapse
Affiliation(s)
- Paula O Cooper
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute for Cancer Research, West Lafayette, IN, USA
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Jiang Yang
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA.
- Purdue Institute for Cancer Research, West Lafayette, IN, USA.
| | - Hsing-Hui Wang
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute for Cancer Research, West Lafayette, IN, USA
- Immune Monitoring and Genomics Facility, Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Meaghan M Broman
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute for Cancer Research, West Lafayette, IN, USA
| | | | | | - Bingyu Yan
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | - Gada D Awdalkreem
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute for Cancer Research, West Lafayette, IN, USA
| | - Gregory M Cresswell
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute for Cancer Research, West Lafayette, IN, USA
- Flow Cytometry Core Facility, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Liang Wang
- Department of Pharmacology and Toxicology, Department of Urology, Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Emery Goossens
- Department of Statistics, Purdue University, West Lafayette, IN, USA
| | - Nadia A Lanman
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute for Cancer Research, West Lafayette, IN, USA
| | - Rebecca W Doerge
- Department of Statistics, Purdue University, West Lafayette, IN, USA
- Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Faye Zheng
- Department of Statistics, Purdue University, West Lafayette, IN, USA
- Sorcero, Inc., Washington, DC, USA
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Department of Surgery (Urology), Brown University Warren Alpert Medical School, the Legorreta Cancer Center at Brown University, and Brown University Health, Providence, RI, USA
| | - Saeed Alqahtani
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute for Cancer Research, West Lafayette, IN, USA
| | - Scott A Crist
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
- Purdue Institute for Cancer Research, West Lafayette, IN, USA
- Carver College of Medicine, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, USA
| | | | - Majid Kazemian
- Purdue Institute for Cancer Research, West Lafayette, IN, USA
- Department of Computer Science, Purdue University, West Lafayette, IN, USA
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | - Travis J Jerde
- Department of Pharmacology and Toxicology, Department of Urology, Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Timothy L Ratliff
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA.
- Purdue Institute for Cancer Research, West Lafayette, IN, USA.
| |
Collapse
|
36
|
Lu Z, Xu L, Wang X. BIT: Bayesian Identification of Transcriptional Regulators from Epigenomics-Based Query Region Sets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.02.597061. [PMID: 38895220 PMCID: PMC11185535 DOI: 10.1101/2024.06.02.597061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Transcriptional regulators (TRs) are master controllers of gene expression and play a critical role in both normal tissue development and disease progression. However, existing computational methods for identification of TRs regulating specific biological processes have significant limitations, such as relying on distance on a linear chromosome or binding motifs that have low specificity. Many also use statistical tests in ways that lack interpretability and rigorous confidence measures. We introduce BIT, a novel Bayesian hierarchical model for in-silico TR identification. Leveraging a comprehensive library of TR ChIP-seq data, BIT offers a fully integrated Bayesian approach to assess genome-wide consistency between user-provided epigenomic profiling data and the TR binding library, enabling the identification of critical TRs while quantifying uncertainty. It avoids estimation and inference in a sequential manner or numerous isolated statistical tests, thereby enhancing accuracy and interpretability. BIT successfully identified critical TRs in perturbation experiments, functionally essential TRs in various cancer types, and cell-type-specific TRs within heterogeneous cell populations, offering deeper biological insights into transcriptional regulation.
Collapse
Affiliation(s)
- Zeyu Lu
- Department of Statistics and Data Science, Moody School of Graduate and Advanced Studies, Southern Methodist University, Dallas, TX, USA
- Department of Mathematics, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Lin Xu
- Quantitative Biomedical Research Center, Peter O’Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pediatrics, Division of Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xinlei Wang
- Department of Mathematics, University of Texas at Arlington, Arlington, TX 76019, USA
- Division of Data Science, College of Science, University of Texas at Arlington, Arlington, TX 76019, USA
| |
Collapse
|
37
|
Miyahira AK, Soule HR. The 30th Annual Prostate Cancer Foundation Scientific Retreat Report. Prostate 2024; 84:1271-1289. [PMID: 39021296 DOI: 10.1002/pros.24768] [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: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND The 30th Annual Prostate Cancer Foundation (PCF) Scientific Retreat was held at the Omni La Costa Resort in Carlsbad, CA, from October 26 to 28, 2023. A hybrid component was included for virtual attendees. METHODS The Annual PCF Scientific Retreat is a leading international scientific conference focused on pioneering, unpublished, and impactful studies across the spectrum of basic through clinical prostate cancer research, as well as research from related fields with significant potential for improving prostate cancer research and patient outcomes. RESULTS The 2023 PCF Retreat concentrated on key areas of research, including: (i) the biology of cancer stem cells and prostate cancer lineage plasticity; (ii) mechanisms of treatment resistance; (iii) emerging AI applications in diagnostic medicine; (iv) analytical and computational biology approaches in cancer research; (v) the role of nerves in prostate cancer; (vi) the biology of prostate cancer bone metastases; (vii) the contribution of ancestry and genomics to prostate cancer disparities; (viii) prostate cancer 3D genomics; (ix) progress in new targets and treatments for prostate cancer; (x) the biology and translational applications of tumor extracellular vesicles; (xi) updates from PCF TACTICAL Award teams; (xii) novel platforms for small molecule molecular glues and binding inhibitors; and (xiii) diversity, equity and inclusion strategies for advancing cancer care equity. CONCLUSIONS This meeting report summarizes the presentations and discussions from the 2023 PCF Scientific Retreat. We hope that sharing this information will deepen our understanding of current and emerging research and drive future advancements in prostate cancer patient care.
Collapse
Affiliation(s)
- Andrea K Miyahira
- Department of Science, Prostate Cancer Foundation, Santa Monica, California, USA
| | - Howard R Soule
- Department of Science, Prostate Cancer Foundation, Santa Monica, California, USA
| |
Collapse
|
38
|
Ludwig ML, Moline D, Horrmann A, Boytim E, Larson G, Arafa AT, Sayeda M, Lozada JR, Bergom HE, Day A, Dasaraju S, Dehm SM, Murugan P, Hwang J, Drake JM, Antonarakis ES. Integrated multi-omics assessment of lineage plasticity in a prostate cancer patient with brain and dural metastases. NPJ Precis Oncol 2024; 8:215. [PMID: 39349591 PMCID: PMC11443004 DOI: 10.1038/s41698-024-00713-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024] Open
Abstract
Metastases to the brain are rare in prostate cancer. Here, we describe a patient with two treatment-emergent metastatic lesions, one to the brain with neuroendocrine prostate cancer (NEPC) histology and one to the dural membrane of adenocarcinoma histology. We performed genomic, transcriptomic, and proteomic characterization of these lesions and the primary tumor to investigate molecular features promoting these metastases. The two metastatic lesions had high genomic similarity, including TP53 mutation and PTEN deletion, with the most striking difference being the additional loss of RB1 in the NEPC lesion. Interestingly, the dural lesion expressed both androgen receptor and neuroendocrine markers, suggesting amphicrine carcinoma (AMPC). When analyzing pioneer transcription factors, the AMPC lesion exhibited elevated FOXA1 activity while the brain NEPC lesion showed elevated HOXC10, NFYB, and OTX2 expression suggesting novel roles in NEPC formation or brain tropism. Our results highlight the utility of performing multi-omic characterization, especially in rare cancer subtypes.
Collapse
Affiliation(s)
- Megan L Ludwig
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - David Moline
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Alec Horrmann
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Ella Boytim
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Gabrianne Larson
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Ali T Arafa
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Masooma Sayeda
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - John R Lozada
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Hannah E Bergom
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Abderrahman Day
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Sandhyarani Dasaraju
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Scott M Dehm
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Department of Urology, University of Minnesota, Minneapolis, MN, USA
| | - Paari Murugan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Justin Hwang
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Justin M Drake
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
- Department of Urology, University of Minnesota, Minneapolis, MN, USA.
| | - Emmanuel S Antonarakis
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
39
|
Dumesic DA, Rasouli MA, Katz JD, Lu GG, Dharanipragada D, Turcu AF, Grogan TR, Flores KE, Magyar CE, Abbott DH, Chazenbalk GD. The Subcutaneous Adipose Microenvironment as a Determinant of Body Fat Development in Polycystic Ovary Syndrome. J Endocr Soc 2024; 8:bvae162. [PMID: 39345868 PMCID: PMC11424691 DOI: 10.1210/jendso/bvae162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Indexed: 10/01/2024] Open
Abstract
Context Adipose steroid metabolism modifies body fat development in polycystic ovary syndrome (PCOS). Objective To determine whether subcutaneous (SC) abdominal adipose aldo-keto reductase 1C3 (AKR1C3; a marker of testosterone generation) is increased in normal-weight women with PCOS vs age- and body mass index (BMI)-matched normoandrogenic ovulatory women (controls) and is related to SC abdominal adipose activator protein-1 (AP-1; a marker of adipocyte differentiation) and/or androgen receptor (AR) protein expression in predicting fat accretion. Design Prospective cohort study. Setting Academic center. Patients Eighteen normal-weight PCOS women; 17 age- and BMI-matched controls. Interventions Circulating hormone/metabolic determinations, intravenous glucose tolerance testing, total body dual-energy x-ray absorptiometry, SC abdominal fat biopsy, immunohistochemistry. Main Outcome Measures Clinical characteristics, hormonal concentrations, body fat distribution, SC adipose AKR1C3, AR, and AP-1 protein expression. Results Women with PCOS had significantly higher serum androgen levels and greater android/gynoid fat mass ratios than controls. SC adipose AKR1C3, AR, and AP-1 protein expressions were comparable between the study groups, but groups differed in correlations. In PCOS women vs controls, SC adipose AKR1C3 protein expression correlated positively with android and gynoid fat masses and negatively with SC adipose AP-1 protein expression. SC adipose AR protein expression correlated negatively with fasting serum free fatty acid and high-density lipoprotein levels. In both study groups, SC adipose AKR1C3 protein expression negatively correlated with serum cortisol levels. Conclusion In normal-weight PCOS women, SC abdominal adipose AKR1C3 protein expression, in combination with intra-adipose AP-1 and AR-dependent events, predicts fat accretion in the presence of physiological cortisol levels.
Collapse
Affiliation(s)
- Daniel A Dumesic
- Department of Obstetrics and Gynecology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Melody A Rasouli
- Department of Obstetrics and Gynecology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jessica D Katz
- Department of Obstetrics and Gynecology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Gwyneth G Lu
- Department of Obstetrics and Gynecology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Devyani Dharanipragada
- Department of Obstetrics and Gynecology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Adina F Turcu
- Division of Metabolism, Endocrinology, Nutrition and Diabetes, University of Michigan, Ann Arbor, MI 48103, USA
| | - Tristan R Grogan
- Department of Medicine Statistics Core, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - Kimberly E Flores
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Clara E Magyar
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - David H Abbott
- Department of Obstetrics and Gynecology, Wisconsin National Primate Research Center, University of Wisconsin, Madison, Madison, WI 53715, USA
| | - Gregorio D Chazenbalk
- Department of Obstetrics and Gynecology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
40
|
Forbes AN, Xu D, Cohen S, Pancholi P, Khurana E. Discovery of therapeutic targets in cancer using chromatin accessibility and transcriptomic data. Cell Syst 2024; 15:824-837.e6. [PMID: 39236711 PMCID: PMC11415227 DOI: 10.1016/j.cels.2024.08.004] [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: 01/07/2022] [Revised: 09/22/2023] [Accepted: 08/08/2024] [Indexed: 09/07/2024]
Abstract
Most cancer types lack targeted therapeutic options, and when first-line targeted therapies are available, treatment resistance is a huge challenge. Recent technological advances enable the use of assay for transposase-accessible chromatin with sequencing (ATAC-seq) and RNA sequencing (RNA-seq) on patient tissue in a high-throughput manner. Here, we present a computational approach that leverages these datasets to identify drug targets based on tumor lineage. We constructed gene regulatory networks for 371 patients of 22 cancer types using machine learning approaches trained with three-dimensional genomic data for enhancer-to-promoter contacts. Next, we identified the key transcription factors (TFs) in these networks, which are used to find therapeutic vulnerabilities, by direct targeting of either TFs or the proteins that they interact with. We validated four candidates identified for neuroendocrine, liver, and renal cancers, which have a dismal prognosis with current therapeutic options.
Collapse
Affiliation(s)
- Andre Neil Forbes
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Duo Xu
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Sandra Cohen
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Priya Pancholi
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ekta Khurana
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; Caryl and Israel Englander Institute for Precision Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
41
|
Shrestha R, Chesner LN, Zhang M, Zhou S, Foye A, Lundberg A, Weinstein AS, Sjöström M, Zhu X, Moreno-Rodriguez T, Li H, Alumkal JJ, Aggarwal R, Small EJ, Lupien M, Quigley DA, Feng FY. An Atlas of Accessible Chromatin in Advanced Prostate Cancer Reveals the Epigenetic Evolution during Tumor Progression. Cancer Res 2024; 84:3086-3100. [PMID: 38990734 DOI: 10.1158/0008-5472.can-24-0890] [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: 03/15/2024] [Revised: 04/16/2024] [Accepted: 07/03/2024] [Indexed: 07/13/2024]
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) is a lethal disease that resists therapy targeting androgen signaling, the primary driver of prostate cancer. mCRPC resists androgen receptor (AR) inhibitors by amplifying AR signaling or by evolving into therapy-resistant subtypes that do not depend on AR. Elucidation of the epigenetic underpinnings of these subtypes could provide important insights into the drivers of therapy resistance. In this study, we produced chromatin accessibility maps linked to the binding of lineage-specific transcription factors (TF) by performing assay for transposase-accessible chromatin sequencing on 70 mCRPC tissue biopsies integrated with transcriptome and whole-genome sequencing. mCRPC had a distinct global chromatin accessibility profile linked to AR function. Analysis of TF occupancy across accessible chromatin revealed 203 TFs associated with mCRPC subtypes. Notably, ZNF263 was identified as a putative prostate cancer TF with a significant impact on gene activity in the double-negative subtype (AR- neuroendocrine-), potentially activating MYC targets. Overall, this analysis of chromatin accessibility in mCRPC provides valuable insights into epigenetic changes that occur during progression to mCRPC. Significance: Integration of a large cohort of transcriptome, whole-genome, and ATAC sequencing characterizes the chromatin accessibility changes in advanced prostate cancer and identifies therapy-resistant prostate cancer subtype-specific transcription factors that modulate oncogenic programs.
Collapse
Affiliation(s)
- Raunak Shrestha
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Lisa N Chesner
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Meng Zhang
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Stanley Zhou
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Adam Foye
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Arian Lundberg
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
- The Institute of Cancer Research and The Royal Marsden Hospital, London, United Kingdom
| | - Alana S Weinstein
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Martin Sjöström
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Xiaolin Zhu
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Thaidy Moreno-Rodriguez
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Urology, University of California, San Francisco, San Francisco, California
| | - Haolong Li
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Joshi J Alumkal
- Division of Hematology and Oncology, University of Michigan Rogel Cancer Center, Ann Arbor, Michigan
| | - Rahul Aggarwal
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Eric J Small
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, San Francisco, California
- Department of Urology, University of California, San Francisco, San Francisco, California
| | - 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, Canada
| | - David A Quigley
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Urology, University of California, San Francisco, San Francisco, California
- Department of Epidemiology & Biostatistics, University of California, San Francisco, San Francisco, California
| | - Felix Y Feng
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, San Francisco, California
- Department of Urology, University of California, San Francisco, San Francisco, California
| |
Collapse
|
42
|
Mu J, Li R, Zheng Y, Lu Y, Ma L, Yin L, Zhang M, Ma W, Chang M, Liu A, Li J, Zhu H, Wang D. Human intermediate prostate cancer stem cells contribute to the initiation and development of prostate adenocarcinoma. Stem Cell Res Ther 2024; 15:296. [PMID: 39256886 PMCID: PMC11389492 DOI: 10.1186/s13287-024-03917-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 09/02/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Intermediate cells are present in the early stages of human prostate development and adenocarcinoma. While primary cells isolated from benign human prostate tissues or tumors exhibit an intermediate phenotype in vitro, they cannot form tumors in vivo unless genetically modified. It is unclear about the stem cell properties and tumorigenicity of intermediate cells. METHODS We developed a customized medium to culture primary human intermediate prostate cells, which were transplanted into male immunodeficient NCG mice to examine tumorigenicity in vivo. We treated the cells with different concentrations of dihydrotestosterone (DHT) and enzalutamide in vitro and surgically castrated the mice after cell transplantation in vivo. Immunostaining, qRT-PCR, RNA sequencing, and western blotting were performed to characterize the cells in tissues and 2D and 3D cultures. RESULTS We found intermediate cells expressing AR+PSA+CK8+CK5+ in the luminal compartment of human prostate adenocarcinoma by immunostaining. We cultured the primary intermediate cells in vitro, which expressed luminal (AR+PSA+CK8+CK18+), basal (CK5+P63+), intermediate (IVL+), and stem cell (CK4+CK13+PSCA+SOX2+) markers. These cells resisted castration in vitro by upregulating the expression of AR, PSA, and proliferation markers KI67 and PCNA. The intermediate cells had high tumorigenicity in vivo, forming tumors in immunodeficient NCG mice in a month without any genetic modification or co-transplantation with embryonic urogenital sinus mesenchyme (UGSM) cells. We named these cells human castration-resistant intermediate prostate cancer stem cells or CriPCSCs and defined the xenograft model as patient primary cell-derived xenograft (PrDX). Human CriPCSCs resisted castration in vitro and in vivo by upregulating AR expression. Furthermore, human CriPCSCs differentiated into amplifying adenocarcinoma cells of luminal phenotype in PrDX tumors in vivo, which can dedifferentiate into CriPCSCs in vitro. CONCLUSIONS Our study identified and established methods for culturing human CriPCSCs, which had high tumorigenicity in vivo without any genetic modification or UGSM co-transplantation. Human CriPCSCs differentiated into amplifying adenocarcinoma cells of luminal phenotype in the fast-growing tumors in vivo, which hold the potential to dedifferentiate into intermediate stem cells. These cells resisted castration by upregulating AR expression. The human CriPCSC and PrDX methods hold significant potential for advancing prostate cancer research and precision medicine.
Collapse
Affiliation(s)
- Jie Mu
- Institute for Translational Medicine, School of Pharmacy, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266071, China
- College of Life Sciences, Qingdao University, Qingdao, 266071, China
| | - Ruizhi Li
- Institute for Translational Medicine, School of Pharmacy, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266071, China
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
| | - Yu Zheng
- Institute for Translational Medicine, School of Pharmacy, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266071, China
- Department of Urology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266011, China
| | - Yi Lu
- Department of Urology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266011, China
| | - Lei Ma
- Institute for Translational Medicine, School of Pharmacy, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266071, China
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
| | - Lin Yin
- Institute for Translational Medicine, School of Pharmacy, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266071, China
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
| | - Miao Zhang
- Institute for Translational Medicine, School of Pharmacy, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266071, China
| | - Wenyu Ma
- Institute for Translational Medicine, School of Pharmacy, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266071, China
| | - Mengjia Chang
- Institute for Translational Medicine, School of Pharmacy, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266071, China
| | - Aihua Liu
- Institute for Translational Medicine, School of Pharmacy, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266071, China.
- College of Life Sciences, Qingdao University, Qingdao, 266071, China.
| | - Jing Li
- Institute for Translational Medicine, School of Pharmacy, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266071, China.
| | - Hai Zhu
- Department of Urology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266011, China.
| | - Dong Wang
- Institute for Translational Medicine, School of Pharmacy, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
43
|
Pati P, Karkampouna S, Bonollo F, Compérat E, Radić M, Spahn M, Martinelli A, Wartenberg M, Kruithof-de Julio M, Rapsomaniki M. Accelerating histopathology workflows with generative AI-based virtually multiplexed tumour profiling. NAT MACH INTELL 2024; 6:1077-1093. [PMID: 39309216 PMCID: PMC11415301 DOI: 10.1038/s42256-024-00889-5] [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: 12/04/2023] [Accepted: 07/29/2024] [Indexed: 09/25/2024]
Abstract
Understanding the spatial heterogeneity of tumours and its links to disease initiation and progression is a cornerstone of cancer biology. Presently, histopathology workflows heavily rely on hematoxylin and eosin and serial immunohistochemistry staining, a cumbersome, tissue-exhaustive process that results in non-aligned tissue images. We propose the VirtualMultiplexer, a generative artificial intelligence toolkit that effectively synthesizes multiplexed immunohistochemistry images for several antibody markers (namely AR, NKX3.1, CD44, CD146, p53 and ERG) from only an input hematoxylin and eosin image. The VirtualMultiplexer captures biologically relevant staining patterns across tissue scales without requiring consecutive tissue sections, image registration or extensive expert annotations. Thorough qualitative and quantitative assessment indicates that the VirtualMultiplexer achieves rapid, robust and precise generation of virtually multiplexed imaging datasets of high staining quality that are indistinguishable from the real ones. The VirtualMultiplexer is successfully transferred across tissue scales and patient cohorts with no need for model fine-tuning. Crucially, the virtually multiplexed images enabled training a graph transformer that simultaneously learns from the joint spatial distribution of several proteins to predict clinically relevant endpoints. We observe that this multiplexed learning scheme was able to greatly improve clinical prediction, as corroborated across several downstream tasks, independent patient cohorts and cancer types. Our results showcase the clinical relevance of artificial intelligence-assisted multiplexed tumour imaging, accelerating histopathology workflows and cancer biology.
Collapse
Affiliation(s)
| | - Sofia Karkampouna
- Urology Research Laboratory, Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Urology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Francesco Bonollo
- Urology Research Laboratory, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Eva Compérat
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Martina Radić
- Urology Research Laboratory, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Martin Spahn
- Department of Urology, Lindenhofspital Bern, Bern, Switzerland
- Department of Urology, University Duisburg-Essen, Essen, Germany
| | - Adriano Martinelli
- IBM Research Europe, Rüschlikon, Switzerland
- ETH Zürich, Zürich, Switzerland
- Biomedical Data Science Center, Lausanne University Hospital, Lausanne, Switzerland
| | - Martin Wartenberg
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Marianna Kruithof-de Julio
- Urology Research Laboratory, Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Urology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Translational Organoid Resource, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Marianna Rapsomaniki
- IBM Research Europe, Rüschlikon, Switzerland
- Biomedical Data Science Center, Lausanne University Hospital, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
44
|
Li F, Tian J, Zhang L, He H, Song D. A multi-omics approach to reveal critical mechanisms of activator protein 1 (AP-1). Biomed Pharmacother 2024; 178:117225. [PMID: 39084078 DOI: 10.1016/j.biopha.2024.117225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024] Open
Abstract
The Activator Protein 1 (AP-1) transcription factor complex plays a pivotal role in the regulation of cancer-related genes, influencing cancer cell proliferation, invasion, migration, angiogenesis, and apoptosis. Composed of multiple subunits, AP-1 has diverse roles across different cancer types and environmental contexts, but its specific mechanisms remain unclear. The advent of multi-omics approaches has shed light on a more comprehensive understanding of AP-1's role and mechanism in gene regulation. This review collates recent genome-wide data on AP-1 and provides an overview of its expression, structure, function, and interaction across different diseases. An examination of these findings can illuminate the intricate nature of AP-1 regulation and its significant involvement in the progression of different diseases. Moreover, we discuss the potential use of AP-1 as a target for individual therapy and explore the various challenges associated with such an approach. Ultimately, this review provides valuable insights into the biology of AP-1 and its potential as a therapeutic target for cancer and disease treatments.
Collapse
Affiliation(s)
- Fei Li
- Clinical Medical Research Center for Women and Children Diseases, Key Laboratory of Birth Defect Prevention and Genetic Medicine of Shandong Health Commission, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan 250014, China; School of Public Health, North China University of Science and Technology, Tangshan 063000, China
| | - Jiaqi Tian
- Clinical Medical Research Center for Women and Children Diseases, Key Laboratory of Birth Defect Prevention and Genetic Medicine of Shandong Health Commission, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan 250014, China
| | - Lin Zhang
- Clinical Medical Research Center for Women and Children Diseases, Key Laboratory of Birth Defect Prevention and Genetic Medicine of Shandong Health Commission, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan 250014, China
| | - Huan He
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Dandan Song
- Clinical Medical Research Center for Women and Children Diseases, Key Laboratory of Birth Defect Prevention and Genetic Medicine of Shandong Health Commission, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan 250014, China.
| |
Collapse
|
45
|
German B, Alaiwi SA, Ho KL, Nanda JS, Fonseca MA, Burkhart DL, Sheahan AV, Bergom HE, Morel KL, Beltran H, Hwang JH, Freedman ML, Lawrenson K, Ellis L. MYBL2 Drives Prostate Cancer Plasticity: Inhibiting Its Transcriptional Target CDK2 for RB1-Deficient Neuroendocrine Prostate Cancer. CANCER RESEARCH COMMUNICATIONS 2024; 4:2295-2307. [PMID: 39113611 PMCID: PMC11368174 DOI: 10.1158/2767-9764.crc-24-0069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 07/05/2024] [Accepted: 08/06/2024] [Indexed: 09/03/2024]
Abstract
Phenotypic plasticity is a recognized mechanism driving therapeutic resistance in patients with prostate cancer. Although underlying molecular causations driving phenotypic plasticity have been identified, therapeutic success is yet to be achieved. To identify putative master regulator transcription factors (MR-TF) driving phenotypic plasticity in prostate cancer, this work utilized a multiomic approach using genetically engineered mouse models of prostate cancer combined with patient data to identify MYB proto-oncogene like 2 (MYBL2) as a significantly enriched transcription factor in prostate cancer exhibiting phenotypic plasticity. Genetic inhibition of Mybl2 using independent murine prostate cancer cell lines representing phenotypic plasticity demonstrated Mybl2 loss significantly decreased in vivo growth as well as cell fitness and repressed gene expression signatures involved in pluripotency and stemness. Because MYBL2 is currently not druggable, a MYBL2 gene signature was employed to identify cyclin-dependent kinase-2 (CDK2) as a potential therapeutic target. CDK2 inhibition phenocopied genetic loss of Mybl2 and significantly decreased in vivo tumor growth associated with enrichment of DNA damage. Together, this work demonstrates MYBL2 as an important MR-TF driving phenotypic plasticity in prostate cancer. Furthermore, high MYBL2 activity identifies prostate cancer that would be responsive to CDK2 inhibition. SIGNIFICANCE Prostate cancers that escape therapy targeting the androgen receptor signaling pathways via phenotypic plasticity are currently untreatable. Our study identifies MYBL2 as a MR-TF in phenotypic plastic prostate cancer and implicates CDK2 inhibition as a novel therapeutic target for this most lethal subtype of prostate cancer.
Collapse
Affiliation(s)
- Beatriz German
- Department of Surgery, Center for Prostate Disease Research, Murtha Cancer Center Research Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland.
- Walter Reed National Military Medical Center, Bethesda, Maryland.
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland.
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.
| | - Sarah A. Alaiwi
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, Connecticut.
| | - Kun-Lin Ho
- Department of Surgery, Center for Prostate Disease Research, Murtha Cancer Center Research Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland.
- Walter Reed National Military Medical Center, Bethesda, Maryland.
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland.
| | - Jagpreet S. Nanda
- Department of Urology, Cedars-Sinai Medical Center, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California.
| | - Marcos A. Fonseca
- Department of Obstetrics and Gynecology and the Women’s Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California.
| | - Deborah L. Burkhart
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| | - Anjali V. Sheahan
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| | - Hannah E. Bergom
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota.
| | - Katherine L. Morel
- South Australian Immunogenomics Cancer Institute, University of Adelaide, Adelaide, Australia.
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| | - Justin H. Hwang
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota.
| | - Matthew L. Freedman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| | - Kate Lawrenson
- Department of Obstetrics and Gynecology and the Women’s Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California.
- Center for Bioinformatics and Functional Genomics, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California.
| | - Leigh Ellis
- Department of Surgery, Center for Prostate Disease Research, Murtha Cancer Center Research Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland.
- Walter Reed National Military Medical Center, Bethesda, Maryland.
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland.
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.
| |
Collapse
|
46
|
Tanaka H, Yoshida S, Aoyama S, Ikeda S, Kunieda J, Ohashi K, Fukuda S, Waseda Y, Tanaka H, Fujii Y. A case of double-negative prostate cancer with BRCA2 mutation and high tumor mutation burden treated sequentially with olaparib and pembrolizumab. IJU Case Rep 2024; 7:395-398. [PMID: 39224671 PMCID: PMC11366435 DOI: 10.1002/iju5.12766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction Double-negative prostate cancer, an androgen receptor-independent prostate cancer without features of neuroendocrine tumors, is refractory to treatment but could be an ideal candidate for individualized treatment. Case presentation An 85-year-old patient with metastatic castration-resistant prostate cancer without prostate-specific antigen progression presented with local recurrence and liver and lung metastases 6 months after orchiectomy and apalutamide. A liver tumor biopsy led to a diagnosis of double-negative prostate cancer. FoundationOne® CDx showed BRCA2 mutation and high tumor mutation burden. Olaparib and pembrolizumab were administered sequentially, and the patient responded to each treatment for 5 months until radiographic progression. Conclusion Sequential use of olaparib and pembrolizumab may be effective for double-negative prostate cancer with BRCA2 mutations and high tumor mutation burden.
Collapse
Affiliation(s)
- Hiroki Tanaka
- Department of UrologyTokyo Medical and Dental UniversityTokyoJapan
| | - Soichiro Yoshida
- Department of UrologyTokyo Medical and Dental UniversityTokyoJapan
| | - Satoru Aoyama
- Department of Precision Cancer MedicineTokyo Medical and Dental UniversityTokyoJapan
| | - Sadakatsu Ikeda
- Department of Precision Cancer MedicineTokyo Medical and Dental UniversityTokyoJapan
| | - Junko Kunieda
- Department of PathologyTokyo Medical and Dental UniversityTokyoJapan
| | - Kenichi Ohashi
- Department of PathologyTokyo Medical and Dental UniversityTokyoJapan
| | - Shohei Fukuda
- Department of UrologyTokyo Medical and Dental UniversityTokyoJapan
| | - Yuma Waseda
- Department of UrologyTokyo Medical and Dental UniversityTokyoJapan
| | - Hajime Tanaka
- Department of UrologyTokyo Medical and Dental UniversityTokyoJapan
| | - Yasuhisa Fujii
- Department of UrologyTokyo Medical and Dental UniversityTokyoJapan
| |
Collapse
|
47
|
Pan S, Yin R, Zhu H, Shen S, Li Z, Liu B. Prostate cancer cancer-associated fibroblasts with stable markers post-androgen deprivation therapy associated with tumor progression and castration resistant prostate cancer. Cancer Sci 2024; 115:2893-2907. [PMID: 38970292 PMCID: PMC11462979 DOI: 10.1111/cas.16267] [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: 03/28/2024] [Revised: 05/30/2024] [Accepted: 06/18/2024] [Indexed: 07/08/2024] Open
Abstract
The specificity and clinical relevance of cancer-associated fibroblasts (CAFs) in prostate cancer (PCa), as well as the effect of androgen deprivation therapy (ADT) on CAFs, remain to be fully elucidated. Using cell lineage diversity and weighted gene co-expression network analysis (WGCNA), we pinpointed a unique CAF signature exclusive to PCa. The specificity of this CAF signature was validated through single-cell RNA sequencing (scRNA-seq), cell line RNA sequencing, and immunohistochemistry. This signature associates CAFs with tumor progression, elevated Gleason scores, and the emergence of castration resistant prostate cancer (CRPC). Using scRNA-seq on collected samples, we demonstrated that the CAF-specific signature is not altered by ADT, maintaining its peak signal output. Identifying a PCa-specific CAF signature and observing signaling changes in CAFs after ADT lay essential groundwork for further PCa studies.
Collapse
Affiliation(s)
- Shen Pan
- Department of Nuclear MedicineShengjing Hospital of China Medical UniversityShenyangChina
- Department of RadiologyShengjing Hospital of China Medical UniversityShenyangChina
| | - Rui Yin
- Department of UrologyShengjing Hospital of China Medical UniversityShenyangChina
| | - Hehe Zhu
- Department of UrologyShengjing Hospital of China Medical UniversityShenyangChina
| | - Siang Shen
- Department of RadiologyShengjing Hospital of China Medical UniversityShenyangChina
| | - Zhenhua Li
- Department of UrologyShengjing Hospital of China Medical UniversityShenyangChina
| | - Bitian Liu
- Department of UrologyShengjing Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
48
|
Zitnik M, Li MM, Wells A, Glass K, Morselli Gysi D, Krishnan A, Murali TM, Radivojac P, Roy S, Baudot A, Bozdag S, Chen DZ, Cowen L, Devkota K, Gitter A, Gosline SJC, Gu P, Guzzi PH, Huang H, Jiang M, Kesimoglu ZN, Koyuturk M, Ma J, Pico AR, Pržulj N, Przytycka TM, Raphael BJ, Ritz A, Sharan R, Shen Y, Singh M, Slonim DK, Tong H, Yang XH, Yoon BJ, Yu H, Milenković T. Current and future directions in network biology. BIOINFORMATICS ADVANCES 2024; 4:vbae099. [PMID: 39143982 PMCID: PMC11321866 DOI: 10.1093/bioadv/vbae099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 05/31/2024] [Accepted: 07/08/2024] [Indexed: 08/16/2024]
Abstract
Summary Network biology is an interdisciplinary field bridging computational and biological sciences that has proved pivotal in advancing the understanding of cellular functions and diseases across biological systems and scales. Although the field has been around for two decades, it remains nascent. It has witnessed rapid evolution, accompanied by emerging challenges. These stem from various factors, notably the growing complexity and volume of data together with the increased diversity of data types describing different tiers of biological organization. We discuss prevailing research directions in network biology, focusing on molecular/cellular networks but also on other biological network types such as biomedical knowledge graphs, patient similarity networks, brain networks, and social/contact networks relevant to disease spread. In more detail, we highlight areas of inference and comparison of biological networks, multimodal data integration and heterogeneous networks, higher-order network analysis, machine learning on networks, and network-based personalized medicine. Following the overview of recent breakthroughs across these five areas, we offer a perspective on future directions of network biology. Additionally, we discuss scientific communities, educational initiatives, and the importance of fostering diversity within the field. This article establishes a roadmap for an immediate and long-term vision for network biology. Availability and implementation Not applicable.
Collapse
Affiliation(s)
- Marinka Zitnik
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, United States
| | - Michelle M Li
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, United States
| | - Aydin Wells
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, IN 46556, United States
- Lucy Family Institute for Data and Society, University of Notre Dame, Notre Dame, IN 46556, United States
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Kimberly Glass
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Deisy Morselli Gysi
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, United States
- Department of Statistics, Federal University of Paraná, Curitiba, Paraná 81530-015, Brazil
- Department of Physics, Northeastern University, Boston, MA 02115, United States
| | - Arjun Krishnan
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | - T M Murali
- Department of Computer Science, Virginia Tech, Blacksburg, VA 24061, United States
| | - Predrag Radivojac
- Khoury College of Computer Sciences, Northeastern University, Boston, MA 02115, United States
| | - Sushmita Roy
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53715, United States
- Wisconsin Institute for Discovery, Madison, WI 53715, United States
| | - Anaïs Baudot
- Aix Marseille Université, INSERM, MMG, Marseille, France
| | - Serdar Bozdag
- Department of Computer Science and Engineering, University of North Texas, Denton, TX 76203, United States
- Department of Mathematics, University of North Texas, Denton, TX 76203, United States
| | - Danny Z Chen
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Lenore Cowen
- Department of Computer Science, Tufts University, Medford, MA 02155, United States
| | - Kapil Devkota
- Department of Computer Science, Tufts University, Medford, MA 02155, United States
| | - Anthony Gitter
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53715, United States
- Morgridge Institute for Research, Madison, WI 53715, United States
| | - Sara J C Gosline
- Biological Sciences Division, Pacific Northwest National Laboratory, Seattle, WA 98109, United States
| | - Pengfei Gu
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Pietro H Guzzi
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, 88100, Italy
| | - Heng Huang
- Department of Computer Science, University of Maryland College Park, College Park, MD 20742, United States
| | - Meng Jiang
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Ziynet Nesibe Kesimoglu
- Department of Computer Science and Engineering, University of North Texas, Denton, TX 76203, United States
- National Center of Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20814, United States
| | - Mehmet Koyuturk
- Department of Computer and Data Sciences, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Jian Ma
- Ray and Stephanie Lane Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, United States
| | - Alexander R Pico
- Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA 94158, United States
| | - Nataša Pržulj
- Department of Computer Science, University College London, London, WC1E 6BT, England
- ICREA, Catalan Institution for Research and Advanced Studies, Barcelona, 08010, Spain
- Barcelona Supercomputing Center (BSC), Barcelona, 08034, Spain
| | - Teresa M Przytycka
- National Center of Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20814, United States
| | - Benjamin J Raphael
- Department of Computer Science, Princeton University, Princeton, NJ 08544, United States
| | - Anna Ritz
- Department of Biology, Reed College, Portland, OR 97202, United States
| | - Roded Sharan
- School of Computer Science, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Yang Shen
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX 77843, United States
| | - Mona Singh
- Department of Computer Science, Princeton University, Princeton, NJ 08544, United States
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, United States
| | - Donna K Slonim
- Department of Computer Science, Tufts University, Medford, MA 02155, United States
| | - Hanghang Tong
- Department of Computer Science, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States
| | - Xinan Holly Yang
- Department of Pediatrics, University of Chicago, Chicago, IL 60637, United States
| | - Byung-Jun Yoon
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX 77843, United States
- Computational Science Initiative, Brookhaven National Laboratory, Upton, NY 11973, United States
| | - Haiyuan Yu
- Department of Computational Biology, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, United States
| | - Tijana Milenković
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, IN 46556, United States
- Lucy Family Institute for Data and Society, University of Notre Dame, Notre Dame, IN 46556, United States
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, United States
| |
Collapse
|
49
|
Zhu X, Farsh T, Vis D, Yu I, Li H, Liu T, Sjöström M, Shrestha R, Kneppers J, Severson T, Zhang M, Lundberg A, Moreno Rodriguez T, Weinstein AS, Foye A, Mehra N, Aggarwal RR, Bergman AM, Small EJ, Lack NA, Zwart W, Quigley DA, van der Heijden MS, Feng FY. Genomic and transcriptomic features of androgen receptor signaling inhibitor resistance in metastatic castration-resistant prostate cancer. J Clin Invest 2024; 134:e178604. [PMID: 39352383 PMCID: PMC11444163 DOI: 10.1172/jci178604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 08/06/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUNDAndrogen receptor signaling inhibitors (ARSIs) have improved outcomes for patients with metastatic castration-resistant prostate cancer (mCRPC), but their clinical benefit is limited by treatment resistance.METHODSTo investigate the mechanisms of ARSI resistance, we analyzed the whole-genome (n = 45) and transcriptome (n = 31) sequencing data generated from paired metastatic biopsies obtained before initiation of first-line ARSI therapy for mCRPC and after radiographic disease progression. We investigated the effects of genetic and pharmacologic modulation of SSTR1 in 22Rv1 cells, a representative mCRPC cell line.RESULTSWe confirmed the predominant role of tumor genetic alterations converging on augmenting androgen receptor (AR) signaling and the increased transcriptional heterogeneity and lineage plasticity during the emergence of ARSI resistance. We further identified amplifications involving a putative enhancer downstream of the AR and transcriptional downregulation of SSTR1, encoding somatostatin receptor 1, in ARSI-resistant tumors. We found that patients with SSTR1-low mCRPC tumors derived less benefit from subsequent ARSI therapy in a retrospective cohort. We showed that SSTR1 was antiproliferative in 22Rv1 cells and that the FDA-approved drug pasireotide suppressed 22Rv1 cell proliferation.CONCLUSIONOur findings expand the knowledge of ARSI resistance and point out actionable next steps, exemplified by potentially targeting SSTR1, to improve patient outcomes.FUNDINGNational Cancer Institute (NCI), NIH; Prostate Cancer Foundation; Conquer Cancer, American Society of Clinical Oncology Foundation; UCSF Benioff Initiative for Prostate Cancer Research; Netherlands Cancer Institute.
Collapse
MESH Headings
- Male
- Humans
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/pathology
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/metabolism
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/drug effects
- Cell Line, Tumor
- Signal Transduction/drug effects
- Transcriptome
- Neoplasm Metastasis
- Receptors, Somatostatin/genetics
- Receptors, Somatostatin/metabolism
- Gene Expression Regulation, Neoplastic/drug effects
- Androgen Receptor Antagonists/pharmacology
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
Collapse
Affiliation(s)
- Xiaolin Zhu
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
- Division of Hematology and Oncology, Department of Medicine, UCSF, San Francisco, California, USA
| | - Tatyanah Farsh
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
- Department of Radiation Oncology, UCSF, San Francisco, California, USA
| | - Daniël Vis
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Ivan Yu
- Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Haolong Li
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
- Department of Radiation Oncology, UCSF, San Francisco, California, USA
| | - Tianyi Liu
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
- Department of Radiation Oncology, UCSF, San Francisco, California, USA
| | - Martin Sjöström
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
- Department of Radiation Oncology, UCSF, San Francisco, California, USA
| | - Raunak Shrestha
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
- Department of Radiation Oncology, UCSF, San Francisco, California, USA
| | - Jeroen Kneppers
- Division of Oncogenomics, Oncode Institute, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Tesa Severson
- Division of Oncogenomics, Oncode Institute, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Meng Zhang
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
- Department of Radiation Oncology, UCSF, San Francisco, California, USA
| | - Arian Lundberg
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
- Department of Radiation Oncology, UCSF, San Francisco, California, USA
| | - Thaidy Moreno Rodriguez
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
- Department of Urology, UCSF, San Francisco, California, USA
| | - Alana S. Weinstein
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
- Department of Radiation Oncology, UCSF, San Francisco, California, USA
| | - Adam Foye
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
- Division of Hematology and Oncology, Department of Medicine, UCSF, San Francisco, California, USA
| | - Niven Mehra
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Rahul R. Aggarwal
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
- Division of Hematology and Oncology, Department of Medicine, UCSF, San Francisco, California, USA
| | - Andries M. Bergman
- Division of Oncogenomics, Oncode Institute, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Eric J. Small
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
- Division of Hematology and Oncology, Department of Medicine, UCSF, San Francisco, California, USA
| | - Nathan A. Lack
- Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Koç University School of Medicine, Istanbul, Turkey
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Wilbert Zwart
- Division of Oncogenomics, Oncode Institute, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - David A. Quigley
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
- Department of Urology, UCSF, San Francisco, California, USA
| | | | - Felix Y. Feng
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
- Department of Radiation Oncology, UCSF, San Francisco, California, USA
| |
Collapse
|
50
|
Liu C, Chen J, Cong Y, Chen K, Li H, He Q, Chen L, Song Y, Xing Y. PROX1 drives neuroendocrine plasticity and liver metastases in prostate cancer. Cancer Lett 2024; 597:217068. [PMID: 38901665 DOI: 10.1016/j.canlet.2024.217068] [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: 03/16/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/22/2024]
Abstract
With the widespread use of anti-androgen therapy, such as abiraterone and enzalutamide, the incidence of neuroendocrine prostate cancer (NEPC) is increasing. NEPC is a lethal form of prostate cancer (PCa), with a median overall survival of less than one year after diagnosis. In addition to the common bone metastases seen in PCa, NEPC exhibits characteristics of visceral metastases, notably liver metastasis, which serves as an indicator of a poor prognosis clinically. Key factors driving the neuroendocrine plasticity of PCa have been identified, yet the underlying mechanism behind liver metastasis remains unclear. In this study, we identified PROX1 as a driver of neuroendocrine plasticity in PCa, responsible for promoting liver metastases. Mechanistically, anti-androgen therapy alleviates transcriptional inhibition of PROX1. Subsequently, elevated PROX1 levels drive both neuroendocrine plasticity and liver-specific transcriptional reprogramming, promoting liver metastases. Moreover, liver metastases in PCa induced by PROX1 depend on reprogrammed lipid metabolism, a disruption that effectively reduces the formation of liver metastases.
Collapse
Affiliation(s)
- Chunyu Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Jiawei Chen
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Yukun Cong
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Kang Chen
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Haoran Li
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Qingliu He
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Liang Chen
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
| | - Yarong Song
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
| | - Yifei Xing
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
| |
Collapse
|