1
|
Li X, Mamouni K, Zhao R, Bai L, Chen Y, Wu Y, Xie ZR, Sautto GA, Liu D, Bowen NJ, Danaher A, Li D, Cook N, Grayson S, Zhu J, Coleman IM, Nelson PS, Bao Q, Zhou J, Osunkoya AO, Kucuk O, Gera L, Wu D. Novel Skp1 inhibitor has potent preclinical efficacy against castration-resistant prostate cancer. Br J Cancer 2025:10.1038/s41416-025-02993-8. [PMID: 40253488 DOI: 10.1038/s41416-025-02993-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 03/01/2025] [Accepted: 03/21/2025] [Indexed: 04/21/2025] Open
Abstract
BACKGROUND Metastatic, castration-resistant prostate cancer (mCRPC) directly contributes to the mortality and morbidity of prostate cancer. It is imperative to identify new molecular targets and discover effective therapeutic agents against lethal mCRPC. METHODS The anticancer activities and mechanism of action of the small-molecule lead compound were investigated in preclinical models of human prostate cancer. Immunohistochemistry was employed to determine the expression of S-phase kinase-associated protein 1 (Skp1) in human prostate tissues. RESULTS GH501 demonstrates nanomolar potency in the NCI-60 human cancer cell panel and multiple mCRPC cell lines with diverse genetic backgrounds, including those resistant to androgen deprivation therapy drugs. Mechanistically, GH501 may bind Skp1 and disrupt the physical interaction between Skp1 and S-phase kinase-associated protein 2 (Skp2) within the Skp1-Cullin1-F-box protein ubiquitin ligase complexes (SCF), thereby affecting multiple oncogenic signals implicated in mCRPC progression, including p21, p27, β-catenin, cyclin D1, enhancer of zeste homolog 2 (EZH2), c-Myc, and survivin. GH501 exhibits excellent in vitro and in vivo safety pharmacology, and GH501 monotherapy effectively inhibits the in vivo growth of cell- and patient-derived xenografts in intraosseous and subcutaneous models. Skp1 expression is significantly increased in human prostate cancer specimens. CONCLUSION These results indicate that interrupting Skp1-Skp2 interaction is an effective approach to target mCRPC and warrant further preclinical development of GH501 as a promising therapeutic candidate.
Collapse
Affiliation(s)
- Xin Li
- Center for Cancer Research and Therapeutic Development and Department of Biological Sciences, Clark Atlanta University, Atlanta, GA, USA
- Molecular Oncology and Biomarkers Program, Georgia Cancer Center; Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Kenza Mamouni
- Molecular Oncology and Biomarkers Program, Georgia Cancer Center; Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Rui Zhao
- Molecular Oncology and Biomarkers Program, Georgia Cancer Center; Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Lijuan Bai
- Molecular Oncology and Biomarkers Program, Georgia Cancer Center; Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yanhua Chen
- Molecular Oncology and Biomarkers Program, Georgia Cancer Center; Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yifei Wu
- School of Electrical and Computer Engineering, College of Engineering, University of Georgia, Athens, GA, USA
| | - Zhong-Ru Xie
- School of Electrical and Computer Engineering, College of Engineering, University of Georgia, Athens, GA, USA
| | - Giuseppe A Sautto
- Florida Research and Innovation Center, Cleveland Clinic, Port St, Lucie, FL, USA
| | | | - Nathan J Bowen
- Center for Cancer Research and Therapeutic Development and Department of Biological Sciences, Clark Atlanta University, Atlanta, GA, USA
| | - Alira Danaher
- Center for Cancer Research and Therapeutic Development and Department of Biological Sciences, Clark Atlanta University, Atlanta, GA, USA
| | - Dehong Li
- Center for Cancer Research and Therapeutic Development and Department of Biological Sciences, Clark Atlanta University, Atlanta, GA, USA
| | - Nicholas Cook
- Center for Cancer Research and Therapeutic Development and Department of Biological Sciences, Clark Atlanta University, Atlanta, GA, USA
| | - Skylar Grayson
- Center for Cancer Research and Therapeutic Development and Department of Biological Sciences, Clark Atlanta University, Atlanta, GA, USA
| | - Jedidiah Zhu
- Center for Cancer Research and Therapeutic Development and Department of Biological Sciences, Clark Atlanta University, Atlanta, GA, USA
| | - Ilsa M Coleman
- Division of Human Biology, Fred Hutchinson Cancer Center, University of Washington, Seattle, WA, USA
| | - Peter S Nelson
- Division of Human Biology, Fred Hutchinson Cancer Center, University of Washington, Seattle, WA, USA
| | - Qichao Bao
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Adeboye O Osunkoya
- Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Omer Kucuk
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Lajos Gera
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, School of Medicine, Aurora, CO, USA.
- MetCure Therapeutics LLC, Atlanta, GA, USA.
| | - Daqing Wu
- Center for Cancer Research and Therapeutic Development and Department of Biological Sciences, Clark Atlanta University, Atlanta, GA, USA.
- Molecular Oncology and Biomarkers Program, Georgia Cancer Center; Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA.
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA.
- MetCure Therapeutics LLC, Atlanta, GA, USA.
| |
Collapse
|
2
|
Weidle UH, Birzele F. Prostate Cancer: De-regulated Circular RNAs With Efficacy in Preclinical In Vivo Models. Cancer Genomics Proteomics 2025; 22:136-165. [PMID: 39993805 PMCID: PMC11880926 DOI: 10.21873/cgp.20494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 11/28/2025] [Accepted: 12/03/2024] [Indexed: 02/26/2025] Open
Abstract
Therapy resistance, including castration-resistance and metastasis, remains a major hurdle in the treatment of prostate cancer. In order to identify novel therapeutic targets and treatment modalities for prostate cancer, we conducted a comprehensive literature search on PubMed to identify de-regulated circular RNAs that influence treatment efficacy in preclinical prostate cancer-related in vivo models. Our analysis identified 49 circular RNAs associated with various processes, including treatment resistance, transmembrane and secreted proteins, transcription factors, signaling cascades, human antigen R, nuclear receptor binding, ubiquitination, metabolism, epigenetics and other target categories. The identified targets and circular RNAs can be further scrutinized through target validation approaches. Down-regulated circular RNAs are candidates for reconstitution therapy, while up-regulated RNAs can be inhibited using small interfering RNA (siRNA), antisense oligonucleotides (ASO) or clustered regularly interspaced short palindromic repeats/CRISPR associated (CRISPR-CAS)-related approaches.
Collapse
Affiliation(s)
- Ulrich H Weidle
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany;
| | - Fabian Birzele
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| |
Collapse
|
3
|
Li C, Cheng D, Li P. Androgen receptor dynamics in prostate cancer: from disease progression to treatment resistance. Front Oncol 2025; 15:1542811. [PMID: 40008000 PMCID: PMC11850250 DOI: 10.3389/fonc.2025.1542811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
Prostate cancer is the most common cancer among men worldwide, especially in those over 65, and is a leading cause of cancer-related mortality. The disease typically advances from an androgen-dependent state to castration-resistant prostate cancer (CRPC), which poses significant treatment challenges. The androgen receptor (AR) on the X chromosome is a central driver in this process, activating genes that govern proliferation and survival. Mutations and amplifications of the AR are closely associated with disease progression and treatment resistance. While traditional therapies such as androgen deprivation therapy (ADT) and AR antagonists like enzalutamide have been effective, resistance persists due to reactivation of AR signaling through mechanisms like ligand-independent activation. Recent research highlights the role of epigenetic modifications in enhancing AR activity and drug resistance. The tumor microenvironment, particularly interactions with cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs), further complicates treatment by promoting aggressive tumor behavior and immune evasion. Future directions include developing next-generation AR antagonists, identifying AR-related biomarkers for personalized therapy, and exploring combinations with immune checkpoint inhibitors. Additionally, basal cell-lumen-derived organoids provide innovative models that can enhance understanding and treatment strategies in prostate cancer.
Collapse
Affiliation(s)
| | | | - Peng Li
- Center for Reproductive Medicine, Shenyang Jinghua Hospital, Shenyang, China
| |
Collapse
|
4
|
Saini T, Srivastava D, Raut R, Mishra P, Misra A. Insulin-Like Growth Factor 2 mRNA-Binding Protein 2 (IGF2BP2) Promotes Castration-Resistant Prostate Cancer Progression by Regulating AR-V7 mRNA Stability. Cancer Rep (Hoboken) 2025; 8:e70096. [PMID: 39948708 PMCID: PMC11825379 DOI: 10.1002/cnr2.70096] [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: 07/09/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND The emergence of constitutively active androgen receptor (AR) splice variant AR-V7 poses a formidable challenge in treating prostate cancer, as it lacks the ligand binding region targeted by androgen-deprivation therapies such as enzalutamide and abiraterone. AR-V7 is critical for castration-resistant prostate cancer (CRPC) development and progression; however, the molecular mechanisms regulating its expression and biological function remain poorly understood. Here, we investigate the role of IGF2BP2 in regulating AR-V7 expression and CRPC progression. METHODS To determine the clinical relevance of IGF2BP2 in CRPC, we analyzed the mRNA expression data for prostate cancer patients available in the Genomic Data Commons (GDC) Data Portal and cBioPortal. Next to investigate the role of IGF2BP2 in regulating AR-V7 expression and enzalutamide resistance, we performed shRNA-mediated IGF2BP2 knockdown and overexpression experiments followed by qRT-PCR, immunoblot, colony-formation, and MTT assays. Finally, we performed RIP-qPCR, actinomycin-D, and IGF2BP2 domain-deletion analysis to study the mechanism by which IGF2BP2 regulates AR-V7 stability, expression, and enzalutamide resistance in CRPC cells. RESULTS Our analysis revealed that IGF2BP2 is upregulated in CRPC patients and its expression positively correlates with increasing Gleason score in patients with CRPC. We demonstrate that IGF2BP2 silencing leads to downregulation of AR-V7 and its downstream target genes without affecting AR levels. Additionally, IGF2BP2 knockdown also enhances the sensitivity of CRPC cells to enzalutamide while overexpression increases AR-V7 expression and confers increased resistance to enzalutamide. Mechanistically, our experiments demonstrate that IGF2BP2 binds to the intronic splicing enhancer (ISE) region of AR-V7, thereby enhancing its mRNA stability. Furthermore, our domain-deletion analysis pinpoints the role of KH3 and KH4 domains of IGF2BP2 in regulating AR-V7 stability and enzalutamide resistance. CONCLUSIONS Taken together, our findings suggest that IGF2BP2 plays a critical role in regulating AR-V7 expression and stability, offering a novel target for developing therapeutic interventions for CRPC.
Collapse
Affiliation(s)
- Taruna Saini
- Department of BiotechnologyIndian Institute of Technology HyderabadKandiSangareddyIndia
| | - Devesh Srivastava
- Department of BiotechnologyIndian Institute of Technology HyderabadKandiSangareddyIndia
| | - Rajnikant Raut
- Department of BiotechnologyIndian Institute of Technology HyderabadKandiSangareddyIndia
| | - Parul Mishra
- Department of Animal BiologySchool of Life Sciences, University of HyderabadHyderabadIndia
| | - Ashish Misra
- Department of BiotechnologyIndian Institute of Technology HyderabadKandiSangareddyIndia
| |
Collapse
|
5
|
Bessot A, Röhl J, Emmerich M, Klotz A, Ravichandran A, Meinert C, Waugh D, McGovern J, Gunter J, Bock N. ECM-mimicking hydrogel models of human adipose tissue identify deregulated lipid metabolism in the prostate cancer-adipocyte crosstalk under antiandrogen therapy. Mater Today Bio 2025; 30:101424. [PMID: 39866784 PMCID: PMC11764633 DOI: 10.1016/j.mtbio.2024.101424] [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: 09/09/2024] [Revised: 12/19/2024] [Accepted: 12/22/2024] [Indexed: 01/28/2025] Open
Abstract
Antiandrogen therapies are effectively used to treat advanced prostate cancer, but eventually cancer adaptation drives unresolved metastatic castration-resistant prostate cancer (mCRPC). Adipose tissue influences metabolic reprogramming in cancer and was proposed as a contributor to therapy resistance. Using extracellular matrix (ECM)-mimicking hydrogel coculture models of human adipocytes and prostate cancer cells, we show that adipocytes from subcutaneous or bone marrow fat have dissimilar responses under the antiandrogen Enzalutamide. We demonstrate that androgen receptor (AR)-dependent cancer cells (LNCaP) are more influenced by human adipocytes than AR-independent cells (C4-2B), with altered lipid metabolism and adipokine secretion. This response changes under Enzalutamide, with increased AR expression and adipogenic and lipogenic genes in cancer cells and decreased lipid content and gene dysregulation associated with insulin resistance in adipocytes. This is in line with the metabolic syndrome that men with mCRPC under Enzalutamide experience. The all-human, all-3D, models presented here provide a significant advance to dissect the role of fat in therapy response for mCRPC.
Collapse
Affiliation(s)
- Agathe Bessot
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia
- Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia
- Max Planck Queensland Centre, Brisbane, QLD, 4000, Australia
- Australian Prostate Cancer Research Centre (APCRC-Q), QUT, Brisbane, QLD, 4102, Australia
| | - Joan Röhl
- Faculty of Health Sciences and Medicine, Bond University, Robina, QLD, 4226, Australia
| | - Maria Emmerich
- School of Computation, Information and Technology, Technical University of Munich (TUM), Munich, Germany
| | - Anton Klotz
- Division of Gastroenterology and Hepatology, Department of Internal Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Akhilandeshwari Ravichandran
- Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia
- School of Mechanical, Medical and Process Engineering, Faculty of Engineering, QUT, Brisbane, QLD 4000, Australia
- Australian Research Council (ARC) Training Centre for Cell and Tissue Engineering Technologies (CTET), QUT, Brisbane, QLD 4000, Australia
| | | | - David Waugh
- Centre for Cancer Biology, University of South Australia, Adelaide, SA 5005, Australia
| | - Jacqui McGovern
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia
- Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia
- Max Planck Queensland Centre, Brisbane, QLD, 4000, Australia
- Australian Research Council (ARC) Training Centre for Cell and Tissue Engineering Technologies (CTET), QUT, Brisbane, QLD 4000, Australia
| | - Jenni Gunter
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia
- Australian Prostate Cancer Research Centre (APCRC-Q), QUT, Brisbane, QLD, 4102, Australia
| | - Nathalie Bock
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia
- Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia
- Max Planck Queensland Centre, Brisbane, QLD, 4000, Australia
- Australian Prostate Cancer Research Centre (APCRC-Q), QUT, Brisbane, QLD, 4102, Australia
- Australian Research Council (ARC) Training Centre for Multiscale 3D Imaging, Modelling, and Manufacturing (M3D Innovation), Queensland University of Technology, Brisbane, QLD 4000, Australia
| |
Collapse
|
6
|
Ali BM, El-Abhar HS, Mohamed G, Nassar HR, Aliedin N, Sharaky M, Shouman SA, Kamel M. A study of the role of androgen receptor and androgen receptor variant 7 in TNBC patients and the effect of their targeting by Enzalutamide and EPI-001 in MDA-MB-231. J Steroid Biochem Mol Biol 2025; 245:106636. [PMID: 39536950 DOI: 10.1016/j.jsbmb.2024.106636] [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: 08/20/2024] [Revised: 11/10/2024] [Accepted: 11/11/2024] [Indexed: 11/16/2024]
Abstract
The lack of targeted therapy for triple-negative breast cancer (TNBC) is among the mainsprings of its poor prognosis. This study aimed to elucidate the role of the androgen receptor (AR) and its splice variant 7 (ARv7) in TNBC patients. Further, the molecular impact of their blockers, Enzalutamide and EPI-001, on the TNBC cell line MDA-MB-231 was investigated. Thereby, immunohistochemical expression of AR/ARv7 was assessed for TNBC Egyptian patients. Moreover, bioinformatics analysis of AR/ARv7 RNA status was carried out on TNBC patients from The Cancer Genome Atlas Breast Carcinoma project (TCGA-BRCA). Data from both groups was correlated with patients' clinicopathological features. Besides, scratch wound healing assay and ELISA were employed to assess the effect of AR/ARv7 blockers on several metastasis markers in MDA-MB-231 cell line. In the Egyptian-TNBC patients, AR expression was associated with worse 7-year DFS (40.6 ± 18.6 %). In addition, ARv7 showed cytoplasmic and nuclear patterns, and both cytoplasmic and nuclear ARv7+ patients demonstrated a worse 7-year DFS (22.7 ± 17.7 % and 20 ± 17.9 %) and overall survival (63.6 ± 14.5 % and 40 ± 21.8 %). Importantly, 80 % of the nuclear ARv7+ patients developed distant metastasis. The data of the TCGA-TNBC patients showed a tendency for poor outcomes in the high ARv7-expressing patients. Molecularly, in MDA-MB-231, both inhibitors modulated metastasis and epithelial to mesenchymal transition (EMT) markers ROCK1, ROCK2, c-Myc, E-cadherin and N-cadherin, with EPI-001 downregulating NF-ĸB level as well. We concluded that ARv7 indicated poor prognosis in the studied cohorts and that blocking of AR/ARv7 abated metastasis and key regulators of EMT in MDA-MB-231, at least in part by targeting ROCK/NF-ĸB/c-Myc axis.
Collapse
Affiliation(s)
- Belal M Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt; Ministry of Health and Population, Egypt.
| | - Hanan S El-Abhar
- Department of Pharmacology, Toxicology, and Biochemistry, Faculty of Pharmacy, Future University in Egypt (FUE), Cairo, Egypt.
| | - Ghada Mohamed
- Department of Pathology, National Cancer Institute, Cairo University, Cairo, Egypt.
| | - Hanan R Nassar
- Department of Medical Oncology, National Cancer Institute, Cairo University, Cairo, Egypt.
| | - Nelly Aliedin
- Department of Medical Statistics Department, National Cancer Institute, Cairo University, Cairo, Egypt.
| | - Marwa Sharaky
- Department of Cancer Biology, Unit of Pharmacology and Experimental Therapeutics, National Cancer Institute, Cairo University, Cairo, Egypt.
| | - Samia A Shouman
- Department of Cancer Biology, Unit of Pharmacology and Experimental Therapeutics, National Cancer Institute, Cairo University, Cairo, Egypt.
| | - Marwa Kamel
- Department of Cancer Biology, Unit of Pharmacology and Experimental Therapeutics, National Cancer Institute, Cairo University, Cairo, Egypt.
| |
Collapse
|
7
|
Ortiz-Hernandez GL, Sanchez-Hernandez ES, Ochoa PT, Casiano CA. The Emerging Roles of the Stress Epigenetic Reader LEDGF/p75 in Cancer Biology and Therapy Resistance: Mechanisms and Targeting Opportunities. Cancers (Basel) 2024; 16:3957. [PMID: 39682146 DOI: 10.3390/cancers16233957] [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: 10/23/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
The lens epithelium derived growth factor of 75 kD (LEDGF/p75) is a transcription co-activator and epigenetic reader that has emerged as a stress oncoprotein in multiple human cancers. Growing evidence indicates that it promotes tumor cell survival against certain therapeutic drugs. The amino (N)-terminal region of LEDGF/p75 contains a PWWP domain that reads methylated histone marks, critical for recognizing transcriptionally active chromatin sites. Its carboxyl (C)-terminus has an integrase binding domain (IBD) that serves as the binding site for the HIV-1 integrase and multiple oncogenic transcription factors. Acting as hubs for protein-protein interactions, both domains facilitate the tethering of oncogenic transcription factors and regulators to active chromatin to regulate mRNA splicing, promote DNA repair, and enhance the expression of stress and cancer-related genes that contribute to tumor cell aggressiveness and chemoresistance. This review summarizes our current knowledge of the emerging roles of LEDGF/p75 in cancer biology and therapy resistance and discusses its potential as a novel oncotherapeutic target in combinatorial treatments.
Collapse
Affiliation(s)
- Greisha L Ortiz-Hernandez
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Evelyn S Sanchez-Hernandez
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Pedro T Ochoa
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Carlos A Casiano
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
- Department of Medicine, Division of Rheumatology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
- Cancer Center, Loma Linda University Health, Loma Linda, CA 92350, USA
| |
Collapse
|
8
|
Srivastava TP, Ajmeriya S, Goel I, Talukdar J, Srivastava A, Parshad R, Deo SVS, Mathur SR, Gogia A, Rai A, Dhar R, Karmakar S. Prognostic role of Androgen Receptor splice variant 7 (AR-V7) in the pathogenesis of breast cancer. BMC Cancer 2024; 24:1398. [PMID: 39538154 PMCID: PMC11562864 DOI: 10.1186/s12885-024-13165-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND The Androgen Receptor (AR) has emerged as an endocrine therapy target in Breast Cancer, exhibiting up to 80% expression in clinical cases. AR-V7, a constitutively activated splice variant of AR with a truncated ligand-binding domain (LBD), demonstrates ligand-independent transcriptional activity and resistance to nonsteroidal antiandrogens like Bicalutamide or Enzalutamide, targeting the LBD. In metastatic prostate cancer, elevated AR-V7 levels lead to therapeutic resistance and increased metastasis. METHODS In this study, we evaluated the expression of AR and AR-V7 in cell lines and a cohort of 89 patients undergoing surgical intervention for treatment-naïve breast cancer. Further clinicopathological correlations and survival analysis were performed to evaluate the relationship between the AR and AR-V7 expression and clinical outcomes. RESULTS AR-V7/AR-FL ratio was elevated in the TNBC cell line and downregulation of AR-FL upon AR antagonists' treatment led to a compensatory increase in AR-V7. Clinical samples showed significantly elevated expression of AR and AR-V7 in tumors compared to control cases. Further clinicopathological correlation revealed aggressive clinical traits, higher pathological grades, and poor survival with AR-V7 expression. CONCLUSIONS Our study unravels AR-V7 as a marker for poor clinical outcomes, predicting breast cancer aggressiveness, and encourages consideration of AR-V7 as a probable target for therapeutic intervention.
Collapse
Affiliation(s)
| | - Swati Ajmeriya
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Isha Goel
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Joyeeta Talukdar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Anurag Srivastava
- Department of Surgical Disciplines, All India Institute of Medical Sciences, New Delhi, India
| | - Rajinder Parshad
- Department of Surgical Disciplines, All India Institute of Medical Sciences, New Delhi, India
| | - S V S Deo
- Department of Surgical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Sandeep R Mathur
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Ajay Gogia
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Avdhesh Rai
- DBT Centre For Molecular Biology and Cancer Research, Dr. Bhubaneswar Borooah Cancer Institute, Guwahati, India
| | - Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
9
|
Marek RD, Halabi S, Wang ME, McBane J, Wei J, Wang T, Yang X, Liu C, Lei G, Lyerly HK, Chen M, Trotter TN, Hartman ZC. Vaccination Against Androgen Receptor Splice Variants to Immunologically Target Prostate Cancer. Vaccines (Basel) 2024; 12:1273. [PMID: 39591176 PMCID: PMC11599078 DOI: 10.3390/vaccines12111273] [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: 09/30/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Background/Objectives: Androgen receptor (AR) expression and signaling are critical for the progression of prostate cancer and have been the therapeutic focus of prostate cancer for over 50 years. While a variety of agents have been developed to target this axis, many of these fail due to the emergent expression of AR RNA splice variants, such as AR-V7, that can signal independently of ligand binding. Other therapies, such as vaccination against prostate-specific antigens, have achieved FDA approvals but have fallen short of being incorporated as standard-of-care therapies for advanced prostate cancer. This may be due to the elevated level of immunosuppression observed in prostate cancer, which remains largely refractory to immune checkpoint blockade. Methods: We developed a vaccine targeting AR-V7, a common isoform associated with treatment resistance, and demonstrated its ability to elicit AR-V7-specific immunity and enable anti-tumor responses against AR-V7+ cancers in subcutaneous tumor models. Results: Our studies also revealed that AR-V7 expression conferred an immune suppressive phenotype that was significant in a non-AR-dependent prostate cancer model. Notably, in this model, we found that vaccination in combination with enzalutamide, an AR antagonist, suppressed these aggressive immune suppressive cancers and resulted in enhanced survival in comparison to control vaccinated and enzalutamide-treated mice. While anti-PD-1 immune checkpoint inhibition (ICI) alone slowed tumor growth, the majority of vaccinated mice that received anti-PD-1 therapy showed complete tumor elimination. Conclusions: Collectively, these results validate the importance of AR signaling in prostate cancer immune suppression and suggest the potential of AR-V7-specific vaccines as therapeutic strategies against prostate cancer, offering significant protective and therapeutic anti-tumor responses, even in the presence of androgen signaling inhibitors.
Collapse
Affiliation(s)
- Robert D. Marek
- Department of Pathology, Duke University, Durham, NC 27710, USA; (R.D.M.); (M.-E.W.)
| | - Selena Halabi
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Mu-En Wang
- Department of Pathology, Duke University, Durham, NC 27710, USA; (R.D.M.); (M.-E.W.)
- Duke Cancer Institute, Duke University, Durham, NC 2771, USA;
| | - Jason McBane
- Department of Surgery, Duke University, Durham, NC 27777, USA; (J.M.); (J.W.); (T.N.T.)
| | - Junping Wei
- Department of Surgery, Duke University, Durham, NC 27777, USA; (J.M.); (J.W.); (T.N.T.)
| | - Tao Wang
- Department of Surgery, Duke University, Durham, NC 27777, USA; (J.M.); (J.W.); (T.N.T.)
| | - Xiao Yang
- Department of Surgery, Duke University, Durham, NC 27777, USA; (J.M.); (J.W.); (T.N.T.)
| | - Congxiao Liu
- Department of Surgery, Duke University, Durham, NC 27777, USA; (J.M.); (J.W.); (T.N.T.)
| | - Gangjun Lei
- Department of Surgery, Duke University, Durham, NC 27777, USA; (J.M.); (J.W.); (T.N.T.)
| | - Herbert Kim Lyerly
- Duke Cancer Institute, Duke University, Durham, NC 2771, USA;
- Department of Surgery, Duke University, Durham, NC 27777, USA; (J.M.); (J.W.); (T.N.T.)
| | - Ming Chen
- Department of Pathology, Duke University, Durham, NC 27710, USA; (R.D.M.); (M.-E.W.)
- Duke Cancer Institute, Duke University, Durham, NC 2771, USA;
| | - Timothy N. Trotter
- Department of Surgery, Duke University, Durham, NC 27777, USA; (J.M.); (J.W.); (T.N.T.)
| | - Zachary C. Hartman
- Department of Pathology, Duke University, Durham, NC 27710, USA; (R.D.M.); (M.-E.W.)
- Duke Cancer Institute, Duke University, Durham, NC 2771, USA;
- Department of Surgery, Duke University, Durham, NC 27777, USA; (J.M.); (J.W.); (T.N.T.)
- Department of Immunobiology, Duke University, Durham, NC 27708, USA
| |
Collapse
|
10
|
Wang R, Pan Y, Zhang L, Wang J, Ni J, Ding Y, Wang S, Yin J, Ding L, Ran X, Fan S, Sun Q, Tan SY, Koeffler HP, Li J, Mi Y, Chen YQ. Prebiotic stachyose inhibits PRDX5 activity and castration-resistant prostate cancer development. Int J Biol Macromol 2024; 278:134844. [PMID: 39168191 DOI: 10.1016/j.ijbiomac.2024.134844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 08/08/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024]
Abstract
Stachyose (STA) is a prebiotic with poor oral bioavailability. In this study, we developed stachyose caproate (C6-STA), as a novel STA derivative, to demonstrate its high adsorption rate via oral administration. Pharmacokinetic analysis reveals that after absorption, the STA derived from C6-STA reaches its highest peak in the blood, liver, and kidney at 20 min, 30 min, and 12-24 h, with approximate levels of 1200 μg/mL, 0.14 μg/mL, and 0.2-0.3 μg/mL, respectively. In addition, the accumulation of STA in prostate tissues of mice with castration-resistant prostate cancer (CRPC) (1.75 μg/mg) is 10-fold higher than that in normal prostate tissues (0.14 μg/mg). The analysis also reveals that C6-STA has t1/2 of 12.8 h and Tmax of 0.25 h, indicating that it has the potential to be used as a promising drug in clinical practice. The toxicological evaluation shows no obvious side effects of C6-STA in mice administered with a 0.2 g/kg intragastric dose. Pharmacodynamic analysis and mechanism investigation of C6-STA show its ability to inhibit peroxiredoxin 5 (PRDX5) enzyme activity, disrupt PRDX5-nuclear factor erythroid 2-related factor 2 (NRF2) interaction, and decrease NAD(P)H quinone dehydrogenase 1 (NQO1) levels. NQO1 decrease further causes the accumulation of quinone radicals, which ultimately leads to the apoptosis of LNCaP cell-derived drug-tolerant persister (DTP) cells and slows CRPC progression. Our study discovered the anti-tumor activity of stachyose and shows that prebiotics have biological functions in vivo besides in the gut. Further investigation of C6-STA, especially in CRPC patients, is warranted.
Collapse
Affiliation(s)
- Rong Wang
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, China; School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Yu Pan
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Lan Zhang
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Jun Wang
- First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China; Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, China
| | - Jiang Ni
- Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, China
| | - Yang Ding
- College of Pharmacy, Pharmaceutical Series, China Pharmaceutical University, Nanjing, China
| | - Shaopeng Wang
- Jiangnan University Medical Center, Jiangnan University, Wuxi, China
| | - Jian Yin
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology & School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China
| | - Lingwen Ding
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore; Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xuebin Ran
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Shuangyi Fan
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Qiaoyang Sun
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Singapore, Singapore
| | - Soo Yong Tan
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - H Phillip Koeffler
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore; Division of Hematology/Oncology, Cedars-Sinai Medical Center, UCLA School of Medicine, CA, Los Angeles, USA
| | - Jie Li
- First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China.
| | - Yuanyuan Mi
- Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, China.
| | - Yong Q Chen
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, China; School of Food Science and Technology, Jiangnan University, Wuxi, China.
| |
Collapse
|
11
|
Cao Y, Jia M, Duan C, Yang Z, Cheng B, Wang R. The m 6A regulators in prostate cancer: molecular basis and clinical perspective. Front Pharmacol 2024; 15:1448872. [PMID: 39268470 PMCID: PMC11391310 DOI: 10.3389/fphar.2024.1448872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/14/2024] [Indexed: 09/15/2024] Open
Abstract
Prostate cancer (PCa) is the second leading cause of cancer-related death among men in western countries. Evidence has indicated the significant role of the androgen receptor (AR) as the main driving factor in controlling the development of PCa, making androgen receptor inhibition (ARI) therapy a pivotal management approach. In addition, AR independent signaling pathways also contribute to PCa progression. One such signaling pathway that has garnered our attention is N6-Methyladenosine (m6A) signaling, which refers to a chemical modification on RNA with crucial roles in RNA metabolism and disease progression, including PCa. It is important to comprehensively summarize the role of each individual m6A regulator in PCa development and understand its interaction with AR signaling. This review aims to provide a thorough summary of the involvement of m6A regulators in PCa development, shedding light on their upstream and downstream signaling pathways. This summary sets the stage for a comprehensive review that would benefit the scientific community and clinical practice by enhancing our understanding of the biology of m6A regulators in the context of PCa.
Collapse
Affiliation(s)
- Yu Cao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Man Jia
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Chunyan Duan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Zhihui Yang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Bo Cheng
- Department of Urology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Ronghao Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| |
Collapse
|
12
|
Andolfi C, Bartolini C, Morales E, Gündoğdu B, Puhr M, Guzman J, Wach S, Taubert H, Aigner A, Eder IE, Handle F, Culig Z. MED12 and CDK8/19 Modulate Androgen Receptor Activity and Enzalutamide Response in Prostate Cancer. Endocrinology 2024; 165:bqae114. [PMID: 39253786 PMCID: PMC11398899 DOI: 10.1210/endocr/bqae114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/16/2024] [Accepted: 09/08/2024] [Indexed: 09/11/2024]
Abstract
Prostate cancer progression is driven by androgen receptor (AR) activity, which is a target for therapeutic approaches. Enzalutamide is an AR inhibitor that prolongs the survival of patients with advanced prostate cancer. However, resistance mechanisms arise and impair its efficacy. One of these mechanisms is the expression of AR-V7, a constitutively active AR splice variant. The Mediator complex is a multisubunit protein that modulates gene expression on a genome-wide scale. MED12 and cyclin-dependent kinase (CDK)8, or its paralog CDK19, are components of the kinase module that regulates the proliferation of prostate cancer cells. In this study, we investigated how MED12 and CDK8/19 influence cancer-driven processes in prostate cancer cell lines, focusing on AR activity and the enzalutamide response. We inhibited MED12 expression and CDK8/19 activity in LNCaP (AR+, enzalutamide-sensitive), 22Rv1 (AR-V7+, enzalutamide-resistant), and PC3 (AR-, enzalutamide-insensitive) cells. Both MED12 and CDK8/19 inhibition reduced cell proliferation in all cell lines, and MED12 inhibition reduced proliferation in the respective 3D spheroids. MED12 knockdown significantly inhibited c-Myc protein expression and signaling pathways. In 22Rv1 cells, it consistently inhibited the AR response, prostate-specific antigen (PSA) secretion, AR target genes, and AR-V7 expression. Combined with enzalutamide, MED12 inhibition additively decreased the AR activity in both LNCaP and 22Rv1 cells. CDK8/19 inhibition significantly decreased PSA secretion in LNCaP and 22Rv1 cells and, when combined with enzalutamide, additively reduced proliferation in 22Rv1 cells. Our study revealed that MED12 and CDK8/19 regulate AR activity and that their inhibition may modulate response to enzalutamide in prostate cancer.
Collapse
Affiliation(s)
- Chiara Andolfi
- Department of Urology, Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Caterina Bartolini
- Department of Urology, Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria
- University of Florence, 50 121 Florence, Italy
| | - Elisa Morales
- Department of Urology, Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Johannes Gutenberg University Mainz, 55122 Mainz, Germany
| | - Büşra Gündoğdu
- Department of Urology, Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Graudate School of Science and Engineering, Yıldız Technical University, 34220 Istanbul, Turkey
| | - Martin Puhr
- Department of Urology, Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Juan Guzman
- Department of Urology and Pediatric Urology, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Sven Wach
- Department of Urology and Pediatric Urology, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Helge Taubert
- Department of Urology and Pediatric Urology, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Achim Aigner
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, University of Leipzig, 04107 Leipzig, Germany
| | - Iris E Eder
- Department of Urology, Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Florian Handle
- Department of Urology, Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Institute of Pathology, Neuropathology & Molecular Pathology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Zoran Culig
- Department of Urology, Division of Experimental Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
13
|
Suzuki A, Sato S, Nakaigawa N, Kishida T, Miyagi Y. Combination of Blood Adiponectin and Leptin Levels Is a Predictor of Biochemical Recurrence in Prostate Cancer Invading the Surrounding Adipose Tissue. Int J Mol Sci 2024; 25:8970. [PMID: 39201655 PMCID: PMC11354761 DOI: 10.3390/ijms25168970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/14/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
Biochemical recurrence is a process that progresses to castration-resistant prostate cancer (CRPC) and prediction of biochemical recurrence is useful in determining early therapeutic intervention and disease treatment. Prostate cancer is surrounded by adipose tissue, which secretes adipokines, affecting cancer progression. This study aimed to investigate the correlation between blood adipokines and CRPC biochemical recurrence. We retrospectively analyzed the clinical data, including preoperative serum adipokine levels, of 99 patients with pT3a pN0 prostate cancer who underwent proctectomy between 2011 and 2019. The primary outcome was biochemical recurrence (prostate-specific antigen: PSA > 0.2). We identified 65 non-recurrences and 34 biochemical recurrences (one progressed to CRPC). The initial PSA level was significantly higher (p = 0.006), but serum adiponectin (p = 0.328) and leptin (p = 0.647) levels and their ratio (p = 0.323) were not significantly different in the biochemical recurrence group compared with the non-recurrence group. In contrast, significantly more biochemical recurrences were observed in the group with adiponectin < 6 μg/mL and Leptin < 4 ng/mL (p = 0.046), initial PSA > 15 ng/mL, clinical Gleason pattern ≥ 4, and positive resection margin. A significant difference was also observed in the multivariate analysis (hazard ratio: 4.04, 95% confidence interval: 1.21-13.5, p = 0.0232). Thus, low preoperative serum adiponectin and high leptin levels were significantly associated with biochemical recurrence in adipose tissue-invasive prostate cancer, suggesting that they may be useful predictors of biochemical recurrence. Further studies with larger cases are needed to increase the validity of this study.
Collapse
Affiliation(s)
- Atsuto Suzuki
- Morphological Analysis Laboratory, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Kanagawa, Japan;
- Department of Urology, Kanagawa Cancer Center, Yokohama 241-8515, Kanagawa, Japan
| | - Shinya Sato
- Morphological Analysis Laboratory, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Kanagawa, Japan;
- Department of Pathology, Kanagawa Cancer Center, Yokohama 241-8515, Kanagawa, Japan
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Kanagawa, Japan
| | - Noboru Nakaigawa
- Department of Urology, Kanagawa Cancer Center, Yokohama 241-8515, Kanagawa, Japan
| | - Takeshi Kishida
- Department of Urology, Kanagawa Cancer Center, Yokohama 241-8515, Kanagawa, Japan
| | - Yohei Miyagi
- Department of Pathology, Kanagawa Cancer Center, Yokohama 241-8515, Kanagawa, Japan
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Kanagawa, Japan
| |
Collapse
|
14
|
Su H, Huang L, Zhou J, Yang G. Prostate cancer stem cells and their targeted therapies. Front Cell Dev Biol 2024; 12:1410102. [PMID: 39175878 PMCID: PMC11338935 DOI: 10.3389/fcell.2024.1410102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 07/23/2024] [Indexed: 08/24/2024] Open
Abstract
Prostate cancer (PCa) is the most common malignancy among men worldwide. Through androgen receptor signaling inhibitor (ARSI) treatment, patients eventually succumb to castration-resistant prostate cancer (CRPC). For this, the prostate cancer stem cells (PCSCs), as a minor population of tumor cells that can promote tumor relapse, ARSI resistance, and disease progression, are gaining attention. Therefore, specific therapy targeting PCSCs has momentum. This study reviewed the identification and characterization of PCSCs and PCSC-based putative biomarkers and summarized their mechanisms of action. We further discussed clinical trials of novel therapeutic interventions focused on PCSC-related pathways, the PCSC microenvironment, cutting-edge miRNA therapy, and immunotherapy approaches from a mechanistic standpoint. This review provides updated insights into PCSC plasticity, identifying new PCSC biomarkers and optimized treatments for patients with advanced PCa.
Collapse
Affiliation(s)
- Huilan Su
- Research Center for Translational Medicine, Cancer Stem Cell Institute, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liqun Huang
- Department of Urology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jianjun Zhou
- Research Center for Translational Medicine, Cancer Stem Cell Institute, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guosheng Yang
- Department of Urology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
15
|
Fei X, Xue JW, Wu JZ, Yang CY, Wang KJ, Ma Q. Promising therapy for neuroendocrine prostate cancer: current status and future directions. Ther Adv Med Oncol 2024; 16:17588359241269676. [PMID: 39131727 PMCID: PMC11311189 DOI: 10.1177/17588359241269676] [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: 04/09/2024] [Accepted: 06/24/2024] [Indexed: 08/13/2024] Open
Abstract
Neuroendocrine prostate cancer (NEPC) is a highly aggressive variant of castration-resistant prostate cancer. It is characterized by low or no expression of the androgen receptor (AR), activation of AR-independent signaling, and increased neuroendocrine phenotype. Most of NEPC is induced by treatment of androgen deprivation therapy and androgen receptor pathway inhibitors (ARPIs). Currently, the treatment of NEPC follows the treatment strategy for small-cell lung cancer, lacking effective drugs and specific treatment options. This review summarizes potential novel targets and therapies for NEPC treatment, including epigenetic regulators (zeste homolog 2 inhibitors, lysine-specific demethylase 1 inhibitors), aurora kinase A inhibitors, poly-ADP-ribose polymerase inhibitors, delta-like ligand 3 targeted therapies, a combination of immunotherapies, etc. Other promising targets and future directions are also discussed in this review. These novel targets and therapies may provide new opportunities for the treatment of NEPC.
Collapse
Affiliation(s)
- Xin Fei
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Jia-Wei Xue
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
- Department of Urology, The First Hospital of Ninghai, Ningbo, China
| | - Ji-zhongrong Wu
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
- Department of Urology, Shengzhou People’s Hospital, Shaoxing, China
| | - Chong-Yi Yang
- Department of Urology, The First Hospital of Ninghai, 142 Taoyuan Middle Road, Yuelong Street, Ninghai county, Ningbo, Zhejiang 315699, China
| | - Ke-Jie Wang
- Comprehensive Genitourinary Cancer Center, The First Affiliated Hospital of Ningbo University, 52, Liuting Street, Haishu District, Ningbo, Zhejiang 315010, China
| | - Qi Ma
- Department of Urology, the First Affiliated Hospital of Ningbo University, 52, Liuting Street, Haishu District,Ningbo, Zhejiang 315010, China
- Comprehensive Genitourinary Cancer Center, The First Affiliated Hospital of Ningbo University, 52, Liuting Street, Haishu District, Ningbo, Zhejiang 315010, China
- Yi-Huan Genitourinary Cancer Group, 52, Liuting Street, Haishu District, Ningbo,Zhejiang 315010, China
| |
Collapse
|
16
|
Rahman M, Akter K, Ahmed KR, Fahim MMH, Aktary N, Park MN, Shin SW, Kim B. Synergistic Strategies for Castration-Resistant Prostate Cancer: Targeting AR-V7, Exploring Natural Compounds, and Optimizing FDA-Approved Therapies. Cancers (Basel) 2024; 16:2777. [PMID: 39199550 PMCID: PMC11352813 DOI: 10.3390/cancers16162777] [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: 07/01/2024] [Revised: 07/29/2024] [Accepted: 08/03/2024] [Indexed: 09/01/2024] Open
Abstract
Castration-resistant prostate cancer (CRPC) remains a significant therapeutic challenge due to its resistance to standard androgen deprivation therapy (ADT). The emergence of androgen receptor splice variant 7 (AR-V7) has been implicated in CRPC progression, contributing to treatment resistance. Current treatments, including first-generation chemotherapy, androgen receptor blockers, radiation therapy, immune therapy, and PARP inhibitors, often come with substantial side effects and limited efficacy. Natural compounds, particularly those derived from herbal medicine, have garnered increasing interest as adjunctive therapeutic agents against CRPC. This review explores the role of AR-V7 in CRPC and highlights the promising benefits of natural compounds as complementary treatments to conventional drugs in reducing CRPC and overcoming therapeutic resistance. We delve into the mechanisms of action underlying the anti-CRPC effects of natural compounds, showcasing their potential to enhance therapeutic outcomes while mitigating the side effects associated with conventional therapies. The exploration of natural compounds offers promising avenues for developing novel treatment strategies that enhance therapeutic outcomes and reduce the adverse effects of conventional CRPC therapies. These compounds provide a safer, more effective approach to managing CRPC, representing a significant advancement in improving patient care.
Collapse
Affiliation(s)
- Muntajin Rahman
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.R.); (K.R.A.); (M.M.H.F.); (N.A.); (M.N.P.)
| | - Khadija Akter
- Department of Plasma Bio Display, Kwangwoon University, Seoul 01897, Republic of Korea;
| | - Kazi Rejvee Ahmed
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.R.); (K.R.A.); (M.M.H.F.); (N.A.); (M.N.P.)
| | - Md. Maharub Hossain Fahim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.R.); (K.R.A.); (M.M.H.F.); (N.A.); (M.N.P.)
| | - Nahida Aktary
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.R.); (K.R.A.); (M.M.H.F.); (N.A.); (M.N.P.)
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.R.); (K.R.A.); (M.M.H.F.); (N.A.); (M.N.P.)
| | - Sang-Won Shin
- Department of Humanities & Social Medicine, School of Korean Medicine, Pusan National University, 49 Busandaehak-ro, Mulgeum-eup, Yangsan-si 50612, Republic of Korea
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.R.); (K.R.A.); (M.M.H.F.); (N.A.); (M.N.P.)
- Department of Plasma Bio Display, Kwangwoon University, Seoul 01897, Republic of Korea;
| |
Collapse
|
17
|
Zhou Y, Zhou F, Xu S, Shi D, Ding D, Wang S, Poongavanam V, Tang K, Liu X, Zhan P. Hydrophobic tagging of small molecules: an overview of the literature and future outlook. Expert Opin Drug Discov 2024; 19:799-813. [PMID: 38825802 DOI: 10.1080/17460441.2024.2360416] [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: 01/04/2024] [Accepted: 05/23/2024] [Indexed: 06/04/2024]
Abstract
INTRODUCTION Hydrophobic tagging (HyT) technology presents a distinct therapeutic strategy diverging from conventional small molecule drugs, providing an innovative approach to drug design. This review aims to provide an overview of the HyT literature and future outlook to offer guidance for drug design. AREAS COVERED In this review, the authors introduce the composition, mechanisms and advantages of HyT technology, as well as summarize the detailed applications of HyT technology in anti-cancer, neurodegenerative diseases (NDs), autoimmune disorders, cardiovascular diseases (CVDs), and other fields. Furthermore, this review discusses key aspects of the future development of HyT molecules. EXPERT OPINION HyT emerges as a highly promising targeted protein degradation (TPD) strategy, following the successful development of proteolysis targeting chimeras (PROTAC) and molecular glue. Based on exploring new avenues, modification of the HyT molecule itself potentially enhances the technology. Improved synthetic pathways and emphasis on pharmacokinetic (PK) properties will facilitate the development of HyT. Furthermore, elucidating the biochemical basis by which the compound's hydrophobic moiety recruits the protein homeostasis network will enable the development of more precise assays that can guide the optimization of the linker and hydrophobic moiety.
Collapse
Affiliation(s)
- Yang Zhou
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Fan Zhou
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Shujing Xu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Dazhou Shi
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Dang Ding
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Shuo Wang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | | | - Kai Tang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| |
Collapse
|
18
|
Nazir SU, Mishra J, Maurya SK, Ziamiavaghi N, Bodas S, Teply BA, Dutta S, Datta K. Deciphering the genetic and epigenetic architecture of prostate cancer. Adv Cancer Res 2024; 161:191-221. [PMID: 39032950 DOI: 10.1016/bs.acr.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Prostate cancer, one of the most frequently diagnosed cancers in men, leads to significant mortality worldwide. Its study is important due to the complexity and diversity in its progression, highlighting the urgent need for improved therapeutic strategies. This chapter probes into the genetic and epigenetic factors influencing prostate cancer progression, underscoring the importance of understanding the disease's molecular fundamentals for the development of targeted therapies. It specifically reviews the role of key genetic mutations in genes such as Androgen Receptor, TP53, SPOP, FOXA1 and PTEN which are crucial for the disease onset and a progression. Furthermore, it examines the impact of epigenetic modifications, including DNA methylation and histone modification, which contribute to the cancer's progression by affecting gene expression and cellular behavior. Further, in this chapter we delve into the underlying signaling mechanism, the advancements in targeting genetic and epigenetic alterations in prostate cancer. These findings have revealed promising targets for therapeutic advancements, aiming to understand and identify promising avenues for future therapies. This chapter improves our current understanding of prostate cancer genetic and epigenetic landscape, emphasizing the necessity of advancing our knowledge to refine and expand treatment options for prostate cancer patients.
Collapse
Affiliation(s)
- Sheeraz Un Nazir
- Department of Biochemistry and Molecular Biology, Massy Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Juhi Mishra
- Department of Biochemistry and Molecular Biology, Massy Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Shailendra Kumar Maurya
- Department of Biochemistry and Molecular Biology, Massy Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Negin Ziamiavaghi
- Department of Biochemistry and Molecular Biology, Massy Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Sanika Bodas
- Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Benjamin A Teply
- Internal Medicine, Division of Oncology & Hematology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Samikshan Dutta
- Department of Biochemistry and Molecular Biology, Massy Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Kaustubh Datta
- Department of Biochemistry and Molecular Biology, Massy Cancer Center, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
19
|
Wang Y, Wu N, Li J, Liang J, Zhou D, Cao Q, Li X, Jiang N. The interplay between autophagy and ferroptosis presents a novel conceptual therapeutic framework for neuroendocrine prostate cancer. Pharmacol Res 2024; 203:107162. [PMID: 38554788 DOI: 10.1016/j.phrs.2024.107162] [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: 01/15/2024] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/02/2024]
Abstract
In American men, the incidence of prostate cancer (PC) is the highest among all types of cancer, making it the second leading cause of mortality associated with cancer. For advanced or metastatic PC, antiandrogen therapies are standard treatment options. The administration of these treatments unfortunately carries the potential risk of inducing neuroendocrine prostate cancer (NEPC). Neuroendocrine differentiation (NED) serves as a crucial indicator of prostate cancer development, encompassing various factors such as phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR), Yes-associated protein 1 (YAP1), AMP-activated protein kinase (AMPK), miRNA. The processes of autophagy and ferroptosis (an iron-dependent form of programmed cell death) play pivotal roles in the regulation of various types of cancers. Clinical trials and preclinical investigations have been conducted on many signaling pathways during the development of NEPC, with the deepening of research, autophagy and ferroptosis appear to be the potential target for regulating NEPC. Due to the dual nature of autophagy and ferroptosis in cancer, gaining a deeper understanding of the developmental programs associated with achieving autophagy and ferroptosis may enhance risk stratification and treatment efficacy for patients with NEPC.
Collapse
Affiliation(s)
- Youzhi Wang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Ning Wu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, Beijing 100191, China
| | - Junbo Li
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Jiaming Liang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Diansheng Zhou
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Qian Cao
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Xuesong Li
- Department of Urology, Peking University First Hospital, Institution of Urology, Peking University, Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, National Urological Cancer Center, Beijing 100034, China.
| | - Ning Jiang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China.
| |
Collapse
|
20
|
Chen H, Dong K, Ding J, Xia J, Qu F, Lan F, Liao H, Qian Y, Huang J, Xu Z, Gu Z, Shi B, Yu M, Cui X, Yu Y. CRISPR genome-wide screening identifies PAK1 as a critical driver of ARSI cross-resistance in prostate cancer progression. Cancer Lett 2024; 587:216725. [PMID: 38364963 DOI: 10.1016/j.canlet.2024.216725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/26/2024] [Accepted: 02/10/2024] [Indexed: 02/18/2024]
Abstract
Next-generation androgen receptor signaling inhibitors (ARSIs), such as enzalutamide (Enza) and darolutamide (Daro), are initially effective for the treatment of advanced prostate cancer (PCa) and castration-resistant prostate cancer (CRPC). However, patients often relapse and develop cross-resistance, which consequently makes drug resistance an inevitable cause of CRPC-related mortality. By conducting a comprehensive analysis of GEO datasets, CRISPR genome-wide screening results, ATAC-seq data, and RNA-seq data, we systemically identified PAK1 as a significant contributor to ARSI cross-resistance due to the activation of the PAK1/RELA/hnRNPA1/AR-V7 axis. Inhibition of PAK1 followed by suppression of NF-κB pathways and AR-V7 expression effectively overcomes ARSI cross-resistance. Our findings indicate that PAK1 represents a promising therapeutic target gene for the treatment of ARSI cross-resistant PCa patients in the clinic. STATEMENT OF SIGNIFICANCE: PAK1 drives ARSI cross-resistance in prostate cancer progression.
Collapse
Affiliation(s)
- Haojie Chen
- Department of Urology, School of Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200092, China; Department of Urology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Keqin Dong
- Department of Urology, School of Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200092, China; Department of Urology, Chinese PLA General Hospital of Central Theater Command, Wuhan, 430064, China
| | - Jie Ding
- Department of Urology, School of Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Jia Xia
- Department of Urology, School of Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Fajun Qu
- Department of Urology, School of Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Fuying Lan
- Department of Urology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Haihong Liao
- Department of Urology, School of Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Yuhang Qian
- Department of Urology, School of Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Jiacheng Huang
- Department of Urology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Zihan Xu
- Department of Urology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Zhengqin Gu
- Department of Urology, School of Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200092, China.
| | - Bowen Shi
- Department of Urology, Huadong Hospital Affiliated to Fudan University, Shanghai, China.
| | - Mingming Yu
- Department of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Xingang Cui
- Department of Urology, School of Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200092, China.
| | - Yongjiang Yu
- Department of Urology, School of Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200092, China.
| |
Collapse
|
21
|
Ma B, Liu D, Zheng M, Wang Z, Zhang D, Jian Y, Ma J, Fan Y, Chen Y, Gao Y, Liu J, Li X, Li L. Development of a Double-Stapled Peptide Stabilizing Both α-Helix and β-Sheet Structures for Degrading Transcription Factor AR-V7. JACS AU 2024; 4:816-827. [PMID: 38425893 PMCID: PMC10900202 DOI: 10.1021/jacsau.3c00795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/08/2024] [Accepted: 01/08/2024] [Indexed: 03/02/2024]
Abstract
Peptide drugs offer distinct advantages in therapeutics; however, their limited stability and membrane penetration abilities hinder their widespread application. One strategy to overcome these challenges is the hydrocarbon peptide stapling technique, which addresses issues such as poor conformational stability, weak proteolytic resistance, and limited membrane permeability. Nonetheless, while peptide stapling has successfully stabilized α-helical peptides, it has shown limited applicability for most β-sheet peptide motifs. In this study, we present the design of a novel double-stapled peptide capable of simultaneously stabilizing both α-helix and β-sheet structures. Our designed double-stapled peptide, named DSARTC, specifically targets the androgen receptor (AR) DNA binding domain and MDM2 as E3 ligase. Serving as a peptide-based PROTAC (proteolysis-targeting chimera), DSARTC exhibits the ability to degrade both the full-length AR and AR-V7. Molecular dynamics simulations and circular dichroism analysis validate the successful constraint of both secondary structures, demonstrating that DSARTC is a "first-in-class" heterogeneous-conformational double-stapled peptide drug candidate. Compared to its linear counterpart, DSARTC displays enhanced stability and an improved cell penetration ability. In an enzalutamide-resistant prostate cancer animal model, DSARTC effectively inhibits tumor growth and reduces the levels of both AR and AR-V7 proteins. These results highlight the potential of DSARTC as a more potent and specific peptide PROTAC for AR-V7. Furthermore, our findings provide a promising strategy for expanding the design of staple peptide-based PROTAC drugs, targeting a wide range of "undruggable" transcription factors.
Collapse
Affiliation(s)
- Bohan Ma
- Department
of Urology, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710049, China
| | - Donghua Liu
- Department
of Urology, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710049, China
| | - Mengjun Zheng
- School
of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Zhe Wang
- Institute
of Bioengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
| | - Dize Zhang
- Department
of Urology, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710049, China
| | - Yanlin Jian
- Department
of Urology, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710049, China
| | - Jian Ma
- Department
of Urology, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710049, China
| | - Yizeng Fan
- Department
of Urology, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710049, China
| | - Yule Chen
- Department
of Urology, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710049, China
| | - Yang Gao
- Department
of Urology, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710049, China
| | - Jing Liu
- Department
of Urology, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710049, China
| | - Xiang Li
- School
of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Lei Li
- Department
of Urology, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710049, China
| |
Collapse
|
22
|
Abdulsamad SA, Naeem AA, Zeng H, He G, Jin X, Alenezi BA, Ai J, Zhang J, Ma H, Rudland PS, Ke Y. Experimental treatment efficacy of dmrFABP5 on prostate cancer singly or in combination with drugs in use. Am J Cancer Res 2024; 14:300-323. [PMID: 38323289 PMCID: PMC10839311 DOI: 10.62347/yppt5752] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/12/2024] [Indexed: 03/17/2025] Open
Abstract
Enzalutamide is a drug used to treat prostate cancer (PC) and docetaxel is a drug for chemotherapeutic treatment of diverse cancer types, including PC. The effectiveness of these drugs in treating castration-resistant prostate cancer (CRPC) is poor and therefore CRPC is still largely incurable. However, the bio-inhibitor of fatty acid-binding protein 5 (FABP5), dmrFABP5, which is a mutant form of FABP5 incapable of binding to fatty acids, has been shown recently to be able to suppress the tumorigenicity and metastasis of cultured CRPC cells. The present study investigated the possible synergistic effect of dmrFABP5 combined with either enzalutamide or docetaxel on suppressing the tumorigenic properties of PC cells, including cell viability, migration, invasion and colony proliferation in soft agar. A highly significant synergistic inhibitory effect on these properties was observed when dmrFABP5 was used in combination with enzalutamide on androgen-responsive PC 22RV1 cells. Moreover, a highly significant synergistic inhibitory effect was also observed when dmrFABP5 was combined with docetaxel, and added to 22RV1 cells and to the highly malignant, androgen-receptor (AR)-negative Du145 cells. DmrFABP5 alone failed to produce any suppressive effect when added to the FABP5-negative cell line LNCaP, although enzalutamide could significantly suppress LNCaP cells when used as a single agent. These synergistic inhibitory effects of dmrFABP5 were produced by interrupting the FABP5-related signal transduction pathway in PC cells. Thus, dmrFABP5 appears to be not only a potential single therapeutic agent, but it may also be used in combination with existing drugs to suppress both AR-positive and AR-negative PC.
Collapse
Affiliation(s)
- Saud A Abdulsamad
- Institute of Urology, West China Hospital, Sichuan UniversityNo. 37 Guo Xue Xiang, Chengdu 610041, Sichuan, China
- Department of Molecular and Clinical Cancer Medicine, Liverpool UniversityCRC Building, No. 200 London Road, Liverpool L3 9TA, UK
| | - Abdulghani A Naeem
- Institute of Urology, West China Hospital, Sichuan UniversityNo. 37 Guo Xue Xiang, Chengdu 610041, Sichuan, China
- Department of Molecular and Clinical Cancer Medicine, Liverpool UniversityCRC Building, No. 200 London Road, Liverpool L3 9TA, UK
| | - Hao Zeng
- Institute of Urology, West China Hospital, Sichuan UniversityNo. 37 Guo Xue Xiang, Chengdu 610041, Sichuan, China
| | - Gang He
- Department of Molecular and Clinical Cancer Medicine, Liverpool UniversityCRC Building, No. 200 London Road, Liverpool L3 9TA, UK
- Sichuan Industrial Institute of Antibiotics, Chengdu UniversityChengdu 610081, Sichuan, China
| | - Xi Jin
- Institute of Urology, West China Hospital, Sichuan UniversityNo. 37 Guo Xue Xiang, Chengdu 610041, Sichuan, China
| | - Bandar A Alenezi
- Department of Molecular and Clinical Cancer Medicine, Liverpool UniversityCRC Building, No. 200 London Road, Liverpool L3 9TA, UK
| | - Jianzhong Ai
- Institute of Urology, West China Hospital, Sichuan UniversityNo. 37 Guo Xue Xiang, Chengdu 610041, Sichuan, China
| | - Jiacheng Zhang
- Department of Molecular and Clinical Cancer Medicine, Liverpool UniversityCRC Building, No. 200 London Road, Liverpool L3 9TA, UK
| | - Hongwen Ma
- Institute of Urology, West China Hospital, Sichuan UniversityNo. 37 Guo Xue Xiang, Chengdu 610041, Sichuan, China
| | - Philip S Rudland
- Department of Biochemistry and Systems BiologyBioscience Building, Crown Street, Liverpool L69 3BX, UK
| | - Youqiang Ke
- Institute of Urology, West China Hospital, Sichuan UniversityNo. 37 Guo Xue Xiang, Chengdu 610041, Sichuan, China
- Department of Molecular and Clinical Cancer Medicine, Liverpool UniversityCRC Building, No. 200 London Road, Liverpool L3 9TA, UK
- Sichuan Industrial Institute of Antibiotics, Chengdu UniversityChengdu 610081, Sichuan, China
| |
Collapse
|
23
|
Jameel M, Fatma H, Nadtochii LA, Siddique HR. Molecular Insight into Prostate Cancer: Preventive Role of Selective Bioactive Molecules. Life (Basel) 2023; 13:1976. [PMID: 37895357 PMCID: PMC10608662 DOI: 10.3390/life13101976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/18/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
Prostate cancer (CaP) is one of the most prevalent male malignancies, accounting for a considerable number of annual mortalities. However, the prompt identification of early-stage CaP often faces delays due to diverse factors, including socioeconomic inequalities. The androgen receptor (AR), in conjunction with various other signaling pathways, exerts a central influence on the genesis, progression, and metastasis of CaP, with androgen deprivation therapy (ADT) serving as the primary therapeutic strategy. Therapeutic modalities encompassing surgery, chemotherapy, hormonal intervention, and radiotherapy have been formulated for addressing early and metastatic CaP. Nonetheless, the heterogeneous tumor microenvironment frequently triggers the activation of signaling pathways, culminating in the emergence of chemoresistance, an aspect to which cancer stem cells (CSCs) notably contribute. Phytochemicals emerge as reservoirs of bioactive agents conferring manifold advantages against human morbidity. Several of these phytochemicals demonstrate potential chemoprotective and chemosensitizing properties against CaP, with selectivity exhibited towards malignant cells while sparing their normal counterparts. In this context, the present review aims to elucidate the intricate molecular underpinnings associated with metastatic CaP development and the acquisition of chemoresistance. Moreover, the contributions of phytochemicals to ameliorating CaP initiation, progression, and chemoresistance are also discussed.
Collapse
Affiliation(s)
- Mohd Jameel
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh 202002, India (H.F.)
| | - Homa Fatma
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh 202002, India (H.F.)
| | - Liudmila A. Nadtochii
- Department of Microbiology, Saint Petersburg State Chemical & Pharmaceutical University, 197022 Saint Petersburg, Russia
| | - Hifzur R. Siddique
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh 202002, India (H.F.)
| |
Collapse
|
24
|
Yu Y, Papukashvili D, Ren R, Rcheulishvili N, Feng S, Bai W, Zhang H, Xi Y, Lu X, Xing N. siRNA-based approaches for castration-resistant prostate cancer therapy targeting the androgen receptor signaling pathway. Future Oncol 2023; 19:2055-2073. [PMID: 37823367 DOI: 10.2217/fon-2023-0227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023] Open
Abstract
Androgen deprivation therapy is a common treatment method for metastatic prostate cancer through lowering androgen levels; however, this therapy frequently leads to the development of castration-resistant prostate cancer (CRPC). This is attributed to the activation of the androgen receptor (AR) signaling pathway. Current treatments targeting AR are often ineffective mostly due to AR gene overexpression and mutations, as well as the presence of splice variants that accelerate CRPC progression. Thus there is a critical need for more specific medication to treat CRPC. Small interfering RNAs have shown great potential as a targeted therapy. This review discusses prostate cancer progression and the role of AR signaling in CRPC, and proposes siRNA-based targeted therapy as a promising strategy for CRPC.
Collapse
Affiliation(s)
- Yanling Yu
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
| | | | - Ruimin Ren
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Department of Urology, Taiyuan, 030032, China
| | | | - Shunping Feng
- Southern University of Science & Technology, Shenzhen, 518000, China
| | - Wenqi Bai
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
| | - Huanhu Zhang
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
| | - Yanfeng Xi
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
| | - Xiaoqing Lu
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
| | - Nianzeng Xing
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
| |
Collapse
|
25
|
Sanchez-Hernandez ES, Ochoa PT, Suzuki T, Ortiz-Hernandez GL, Unternaehrer JJ, Alkashgari HR, Diaz Osterman CJ, Martinez SR, Chen Z, Kremsky I, Wang C, Casiano CA. Glucocorticoid Receptor Regulates and Interacts with LEDGF/p75 to Promote Docetaxel Resistance in Prostate Cancer Cells. Cells 2023; 12:2046. [PMID: 37626856 PMCID: PMC10453226 DOI: 10.3390/cells12162046] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/31/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Patients with advanced prostate cancer (PCa) invariably develop resistance to anti-androgen therapy and taxane-based chemotherapy. Glucocorticoid receptor (GR) has been implicated in PCa therapy resistance; however, the mechanisms underlying GR-mediated chemoresistance remain unclear. Lens epithelium-derived growth factor p75 (LEDGF/p75, also known as PSIP1 and DFS70) is a glucocorticoid-induced transcription co-activator implicated in cancer chemoresistance. We investigated the contribution of the GR-LEDGF/p75 axis to docetaxel (DTX)-resistance in PCa cells. GR silencing in DTX-sensitive and -resistant PCa cells decreased LEDGF/p75 expression, and GR upregulation in enzalutamide-resistant cells correlated with increased LEDGF/p75 expression. ChIP-sequencing revealed GR binding sites in the LEDGF/p75 promoter. STRING protein-protein interaction analysis indicated that GR and LEDGF/p75 belong to the same transcriptional network, and immunochemical studies demonstrated their co-immunoprecipitation and co-localization in DTX-resistant cells. The GR modulators exicorilant and relacorilant increased the sensitivity of chemoresistant PCa cells to DTX-induced cell death, and this effect was more pronounced upon LEDGF/p75 silencing. RNA-sequencing of DTX-resistant cells with GR or LEDGF/p75 knockdown revealed a transcriptomic overlap targeting signaling pathways associated with cell survival and proliferation, cancer, and therapy resistance. These studies implicate the GR-LEDGF/p75 axis in PCa therapy resistance and provide a pre-clinical rationale for developing novel therapeutic strategies for advanced PCa.
Collapse
Affiliation(s)
- Evelyn S. Sanchez-Hernandez
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (E.S.S.-H.); (T.S.); (G.L.O.-H.); (J.J.U.); (H.R.A.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (Z.C.); (I.K.); (C.W.)
| | - Pedro T. Ochoa
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (E.S.S.-H.); (T.S.); (G.L.O.-H.); (J.J.U.); (H.R.A.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (Z.C.); (I.K.); (C.W.)
| | - Tise Suzuki
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (E.S.S.-H.); (T.S.); (G.L.O.-H.); (J.J.U.); (H.R.A.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (Z.C.); (I.K.); (C.W.)
| | - Greisha L. Ortiz-Hernandez
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (E.S.S.-H.); (T.S.); (G.L.O.-H.); (J.J.U.); (H.R.A.)
| | - Juli J. Unternaehrer
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (E.S.S.-H.); (T.S.); (G.L.O.-H.); (J.J.U.); (H.R.A.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (Z.C.); (I.K.); (C.W.)
| | - Hossam R. Alkashgari
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (E.S.S.-H.); (T.S.); (G.L.O.-H.); (J.J.U.); (H.R.A.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (Z.C.); (I.K.); (C.W.)
- Department of Physiology, College of Medicine, University of Jeddah, Jeddah 23890, Saudi Arabia
| | - Carlos J. Diaz Osterman
- Department of Basic Sciences, Ponce Health Sciences University, Ponce, PR 00716, USA; (C.J.D.O.); (S.R.M.)
| | - Shannalee R. Martinez
- Department of Basic Sciences, Ponce Health Sciences University, Ponce, PR 00716, USA; (C.J.D.O.); (S.R.M.)
| | - Zhong Chen
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (Z.C.); (I.K.); (C.W.)
- Center for Genomics, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Isaac Kremsky
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (Z.C.); (I.K.); (C.W.)
- Center for Genomics, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Charles Wang
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (Z.C.); (I.K.); (C.W.)
- Center for Genomics, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Carlos A. Casiano
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (E.S.S.-H.); (T.S.); (G.L.O.-H.); (J.J.U.); (H.R.A.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (Z.C.); (I.K.); (C.W.)
- Rheumatology Division, Department of Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| |
Collapse
|
26
|
Maddeboina K, Jonnalagadda SK, Morsy A, Duan L, Chhonker YS, Murry DJ, Penning TM, Trippier PC. Aldo-Keto Reductase 1C3 Inhibitor Prodrug Improves Pharmacokinetic Profile and Demonstrates In Vivo Efficacy in a Prostate Cancer Xenograft Model. J Med Chem 2023; 66:9894-9915. [PMID: 37428858 PMCID: PMC11963376 DOI: 10.1021/acs.jmedchem.3c00732] [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] [Indexed: 07/12/2023]
Abstract
Aldo-keto reductase 1C3 (AKR1C3) is overexpressed in castration-resistant prostate cancer where it acts to drive proliferation and aggressiveness by producing androgens. The reductive action of the enzyme leads to chemoresistance development against various clinical antineoplastics across a range of cancers. Herein, we report the continued optimization of selective AKR1C3 inhibitors and the identification of 5r, a potent AKR1C3 inhibitor (IC50 = 51 nM) with >1216-fold selectivity for AKR1C3 over closely related isoforms. Due to the cognizance of the poor pharmacokinetics associated with free carboxylic acids, a methyl ester prodrug strategy was pursued. The prodrug 4r was converted to free acid 5r in vitro in mouse plasma and in vivo. The in vivo pharmacokinetic evaluation revealed an increase in systemic exposure and increased the maximum 5r concentration compared to direct administration of the free acid. The prodrug 4r demonstrated a dose-dependent effect to reduce the tumor volume of 22Rv1 prostate cancer xenografts without observed toxicity.
Collapse
Affiliation(s)
- Krishnaiah Maddeboina
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
| | - Sravan K Jonnalagadda
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
| | - Ahmed Morsy
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
| | - Ling Duan
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Yashpal S Chhonker
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
| | - Daryl J Murry
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
| | - Trevor M Penning
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Paul C Trippier
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
- UNMC Center for Drug Discovery, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
| |
Collapse
|
27
|
Xu P, Yang JC, Ning S, Chen B, Nip C, Wei Q, Liu L, Johnson OT, Gao AC, Gestwicki JE, Evans CP, Liu C. Allosteric inhibition of HSP70 in collaboration with STUB1 augments enzalutamide efficacy in antiandrogen resistant prostate tumor and patient-derived models. Pharmacol Res 2023; 189:106692. [PMID: 36773708 PMCID: PMC10162009 DOI: 10.1016/j.phrs.2023.106692] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/30/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023]
Abstract
Ubiquitin proteasome activity is suppressed in enzalutamide resistant prostate cancer cells, and the heat shock protein 70/STIP1 homology and U-box-containing protein 1 (HSP70/STUB1) machinery are involved in androgen receptor (AR) and AR variant protein stabilization. Targeting HSP70 could be a viable strategy to overcome resistance to androgen receptor signaling inhibitor (ARSI) in advanced prostate cancer. Here, we showed that a novel HSP70 allosteric inhibitor, JG98, significantly suppressed drug-resistant C4-2B MDVR and CWR22Rv1 cell growth, and enhanced enzalutamide treatment. JG98 also suppressed cell growth in conditional reprogramed cell cultures (CRCs) and organoids derived from advanced prostate cancer patient samples. Mechanistically, JG98 degraded AR/AR-V7 expression in resistant cells and promoted STUB1 nuclear translocation to bind AR-V7. Knockdown of the E3 ligase STUB1 significantly diminished the anticancer effects and partially restored AR-V7 inhibitory effects of JG98. JG231, a more potent analog developed from JG98, effectively suppressed the growth of the drug-resistant prostate cancer cells, CRCs, and organoids. Notably, the combination of JG231 and enzalutamide synergistically inhibited AR/AR-V7 expression and suppressed CWR22Rv1 xenograft tumor growth. Inhibition of HSP70 using novel small-molecule inhibitors coordinates with STUB1 to regulate AR/AR-V7 protein stabilization and ARSI resistance.
Collapse
Affiliation(s)
- Pengfei Xu
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Joy C Yang
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Shu Ning
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Bo Chen
- Department of Urologic Surgery, University of California, Davis, CA, USA; Department of Urology, West China Hospital, Sichuan University, Sichuan, China
| | - Christopher Nip
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Qiang Wei
- Department of Urology, West China Hospital, Sichuan University, Sichuan, China
| | - Liangren Liu
- Department of Urology, West China Hospital, Sichuan University, Sichuan, China
| | - Oleta T Johnson
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Allen C Gao
- Department of Urologic Surgery, University of California, Davis, CA, USA; University of California, Davis Comprehensive Cancer Center, CA, USA
| | - Jason E Gestwicki
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Christopher P Evans
- Department of Urologic Surgery, University of California, Davis, CA, USA; University of California, Davis Comprehensive Cancer Center, CA, USA
| | - Chengfei Liu
- Department of Urologic Surgery, University of California, Davis, CA, USA; University of California, Davis Comprehensive Cancer Center, CA, USA.
| |
Collapse
|
28
|
Sakellakis M. Niclosamide in prostate cancer: An inhibitor of AR-V7, a mitochondrial uncoupler, or more? Cancer Treat Res Commun 2023; 35:100685. [PMID: 36706514 DOI: 10.1016/j.ctarc.2023.100685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/23/2023]
Abstract
A recent phase Ib study investigating the use of reformulated niclosamide in combination with abiraterone and prednisone in patients with castration-resistant prostate cancer (CRPC) demonstrated encouraging preliminary efficacy with low toxicity. Preclinical studies have reported that niclosamide at clinically relevant concentrations inhibits androgen receptor splice variant 7 (AR-V7), a known tumor driver in CRPC. However, the magnitude of anti-tumor effects of niclosamide either used alone or in combination with abiraterone in these experimental models, far exceeded what could have been explained as a simple AR-V7 inhibition. Niclosamide at clinically relevant concentrations also acts as an oxidative phosphorylation (OxPhos) uncoupler in mitochondria. This raises the question whether the observed effects of niclosamide were partly mediated by OxPhos inhibition. Most OxPhos inhibitors did not demonstrate selectivity towards cancer cells and failed to enter clinical practice due to unacceptable toxicity. However, some mitochondrial uncouplers have greater cytotoxicity against cancerous cells compared to non-cancerous. Hyperpolarization of cancer cell mitochondria, or the more alkaline mitochondrial matrix of cancer cells could be potential reasons for this. Niclosamide can also alter Wnt/β-catenin, mTOR, Notch, NF-kB and STAT3 signaling pathways. Hence, the mechanism of action of reformulated niclosamide in CRPC patients requires further investigation. This will potentially lead to new opportunities to develop and investigate even more selective and effective treatments against prostate cancer.
Collapse
Affiliation(s)
- Minas Sakellakis
- Hellenic GU Cancer Group, Athens, Greece; Department of Medical Oncology, Metropolitan Hospital, Athens, 18547, Greece.
| |
Collapse
|