51
|
Oseni SO, Naar C, Pavlović M, Asghar W, Hartmann JX, Fields GB, Esiobu N, Kumi-Diaka J. The Molecular Basis and Clinical Consequences of Chronic Inflammation in Prostatic Diseases: Prostatitis, Benign Prostatic Hyperplasia, and Prostate Cancer. Cancers (Basel) 2023; 15:3110. [PMID: 37370720 DOI: 10.3390/cancers15123110] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 05/23/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Chronic inflammation is now recognized as one of the major risk factors and molecular hallmarks of chronic prostatitis, benign prostatic hyperplasia (BPH), and prostate tumorigenesis. However, the molecular mechanisms by which chronic inflammation signaling contributes to the pathogenesis of these prostate diseases are poorly understood. Previous efforts to therapeutically target the upstream (e.g., TLRs and IL1-Rs) and downstream (e.g., NF-κB subunits and cytokines) inflammatory signaling molecules in people with these conditions have been clinically ambiguous and unsatisfactory, hence fostering the recent paradigm shift towards unraveling and understanding the functional roles and clinical significance of the novel and relatively underexplored inflammatory molecules and pathways that could become potential therapeutic targets in managing prostatic diseases. In this review article, we exclusively discuss the causal and molecular drivers of prostatitis, BPH, and prostate tumorigenesis, as well as the potential impacts of microbiome dysbiosis and chronic inflammation in promoting prostate pathologies. We specifically focus on the importance of some of the underexplored druggable inflammatory molecules, by discussing how their aberrant signaling could promote prostate cancer (PCa) stemness, neuroendocrine differentiation, castration resistance, metabolic reprogramming, and immunosuppression. The potential contribution of the IL1R-TLR-IRAK-NF-κBs signaling molecules and NLR/inflammasomes in prostate pathologies, as well as the prospective benefits of selectively targeting the midstream molecules in the various inflammatory cascades, are also discussed. Though this review concentrates more on PCa, we envision that the information could be applied to other prostate diseases. In conclusion, we have underlined the molecular mechanisms and signaling pathways that may need to be targeted and/or further investigated to better understand the association between chronic inflammation and prostate diseases.
Collapse
Affiliation(s)
- Saheed Oluwasina Oseni
- Department of Biological Sciences, Florida Atlantic University, Boca Raton, FL 33431, USA
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Corey Naar
- Department of Biological Sciences, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Mirjana Pavlović
- Department of Computer and Electrical Engineering, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Waseem Asghar
- Department of Computer and Electrical Engineering, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - James X Hartmann
- Department of Biological Sciences, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Gregg B Fields
- Department of Chemistry & Biochemistry, and I-HEALTH, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Nwadiuto Esiobu
- Department of Biological Sciences, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - James Kumi-Diaka
- Department of Biological Sciences, Florida Atlantic University, Boca Raton, FL 33431, USA
| |
Collapse
|
52
|
Pitzen SP, Dehm SM. Basal epithelial cells in prostate development, tumorigenesis, and cancer progression. Cell Cycle 2023; 22:1303-1318. [PMID: 37098827 PMCID: PMC10228417 DOI: 10.1080/15384101.2023.2206502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 04/27/2023] Open
Abstract
The prostate epithelium is composed of two predominant cell populations: luminal and basal epithelial cells. Luminal cells have a secretory function that supports male fertility while basal cells function in regeneration and maintenance of epithelial tissue. Recent studies in humans and mice have expanded our knowledge of the role and regulation of luminal and basal cells in prostate organogenesis, development, and homeostasis. The insights from healthy prostate biology can inform studies focused on the origins of prostate cancer, progression of the disease, and development of resistance to targeted hormonal therapies. In this review, we discuss a critical role for basal cells in the development and maintenance of healthy prostate tissue. Additionally, we provide evidence supporting a role for basal cells in oncogenesis and therapeutic resistance mechanisms of prostate cancer. Finally, we describe basal cell regulators that may promote lineage plasticity and basal cell identity in prostate cancers that have developed therapeutic resistance. These regulators could serve as therapeutic targets to inhibit or delay resistance and thereby improve outcomes for prostate cancer patients.
Collapse
Affiliation(s)
- Samuel P. Pitzen
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Graduate Program in Molecular, Cellular, and Developmental Biology and Genetics, University of Minnesota, Minneapolis, MN, USA
| | - Scott M. Dehm
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, USA
- Department of Urology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
53
|
Huang Z, Tang Y, Wei Y, Qian J, Kang Y, Wang D, Xu M, Nie L, Chen X, Chen N, Zhou Q. Prognostic Significance of Chromogranin A Expression in the Initial and Second Biopsies in Metastatic Prostate Cancer. J Clin Med 2023; 12:jcm12103362. [PMID: 37240468 DOI: 10.3390/jcm12103362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/27/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
Neuroendocrine differentiation (NED) characterized by the expression of neuroendocrine markers, such as chromogranin A (CgA), is frequently observed in advanced prostate cancer (PCa), the prognostic significance of which is still controversial. Here we specifically addressed the issue of the potential prognostic value of CgA expression in advanced-stage PCa patients with distant metastases and its change over time from metastatic hormone-sensitive (mHSPC) to metastatic castration-resistant prostate cancer (mCRPC). CgA expression was assessed immunohistochemically in initial biopsies of mHSPC, as well as in second biopsies of mCRPC in sixty-eight patients, and its correlation with prognosis (together with conventional clinicopathologic parameters) was analyzed using the Kaplan-Meier method and Cox proportional hazard model. We found that CgA expression was an independent adverse prognostic factor for both mHSPC (CgA positivity ≥ 1%, HR = 2.16, 95% CI: 1.04-4.26, p = 0.031) and mCRPC (CgA ≥ 10%, HR = 20.19, 95% CI: 3.04-329.9, p = 0.008). CgA positivity generally increased from mHSPC to mCRPC and was a negative prognosticator. The assessment of CgA expression may help with the clinical evaluation of advanced-stage patients with distant metastases.
Collapse
Affiliation(s)
- Zhuo Huang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ying Tang
- Department of Pathology, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| | - Yuyan Wei
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingyu Qian
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yifan Kang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Duohao Wang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Miao Xu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ling Nie
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xueqin Chen
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ni Chen
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiao Zhou
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
54
|
Han H, Li H, Ma Y, Zhao Z, An Q, Zhao J, Shi C. Monoamine oxidase A (MAOA): A promising target for prostate cancer therapy. Cancer Lett 2023; 563:216188. [PMID: 37076041 DOI: 10.1016/j.canlet.2023.216188] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/08/2023] [Accepted: 04/11/2023] [Indexed: 04/21/2023]
Abstract
Monoamine oxidase A (MAOA) is a mitochondrial enzyme that catalyzes the oxidative deamination of monoamine neurotransmitters and dietary amines. Previous studies have shown that MAOA is clinically associated with prostate cancer (PCa) progression and plays a key role in almost each stage of PCa, including castrate-resistant prostate cancer, neuroendocrine prostate cancer, metastasis, drug resistance, stemness, and perineural invasion. Moreover, MAOA expression is upregulated not only in cancer cells but also in stromal cells, intratumoral T cells, and tumor-associated macrophages; thus, targeting MAOA can be a multi-pronged approach to disrupt tumor promoting interactions between PCa cells and tumor microenvironment. Furthermore, targeting MAOA can disrupt the crosstalk between MAOA and the androgen receptor (AR) to restore enzalutamide sensitivity, blocks glucocorticoid receptor (GR)- and AR-dependent PCa cell growth, and is a potential strategy for immune checkpoint inhibition, thereby alleviating immune suppression and enhancing T cell immunity-based cancer immunotherapy. MAOA is a promising target for PCa therapy, which deserves further exploration in preclinical and clinical settings.
Collapse
Affiliation(s)
- Hao Han
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; School of Basic Medical Sciences, Medical College of Yan'an University, 580 Bao-Ta Street, Yan'an, Shaanxi, 716000, China
| | - Hui Li
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; School of Basic Medical Sciences, Medical College of Yan'an University, 580 Bao-Ta Street, Yan'an, Shaanxi, 716000, China
| | - Yifan Ma
- Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, 730030, China
| | - Zhite Zhao
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, 710032, China
| | - Qingling An
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Jumei Zhao
- School of Basic Medical Sciences, Medical College of Yan'an University, 580 Bao-Ta Street, Yan'an, Shaanxi, 716000, China.
| | - Changhong Shi
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
55
|
Huna A, Martin N, Bernard D. The senescence-associated secretory phenotype induces neuroendocrine transdifferentiation. Aging (Albany NY) 2023; 15:2819-2821. [PMID: 37071013 PMCID: PMC10188343 DOI: 10.18632/aging.204669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/16/2023] [Indexed: 04/19/2023]
Affiliation(s)
- Anda Huna
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Equipe Labellisée la Ligue Contre le Cancer, France
| | - Nadine Martin
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Equipe Labellisée la Ligue Contre le Cancer, France
| | - David Bernard
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Equipe Labellisée la Ligue Contre le Cancer, France
| |
Collapse
|
56
|
Asai S, Goto Y, Tanigawa K, Tomioka Y, Kato M, Mizuno K, Sakamoto S, Seki N. MiR-15b-5p inhibits castration-resistant growth of prostate cancer cells by targeting the muscarinic cholinergic receptor CHRM3. FEBS Lett 2023; 597:1164-1175. [PMID: 36754848 DOI: 10.1002/1873-3468.14598] [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: 12/02/2022] [Revised: 01/30/2023] [Accepted: 01/30/2023] [Indexed: 02/10/2023]
Abstract
Cholinergic receptor muscarinic 3 (CHRM3)-mediated focal adhesion kinase/YES-associated protein (YAP) signalling is essential for the growth of castration-resistant prostate cancer (CRPC) cells. Here, we evaluated the molecular mechanisms through which CHRM3 overexpression facilitates castration-resistant growth. Small RNA sequencing combined with in silico analyses revealed that CHRM3 was a putative target of miR-15b-5p. Notably, androgen deprivation suppressed miR-15b-5p expression and increased CHRM3 expression. Moreover, miR-15b-5p bound directly to CHRM3 and inhibited YAP activation induced by CHRM3 stimulation. Furthermore, miR-15b-5p abolished the growth of CRPC cells induced by CHRM3 stimulation. We conclude that the miR-15b-5p/CHRM3/YAP signalling axis promotes the castration-resistant growth of prostate cancer.
Collapse
Affiliation(s)
- Shunichi Asai
- Department of Functional Genomics, Chiba University Graduate School of Medicine, Japan
| | - Yusuke Goto
- Department of Functional Genomics, Chiba University Graduate School of Medicine, Japan.,Department of Urology, Chiba University Graduate School of Medicine, Japan
| | - Kengo Tanigawa
- Department of Pulmonary Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Japan
| | - Yuya Tomioka
- Department of Pulmonary Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Japan
| | - Mayuko Kato
- Department of Functional Genomics, Chiba University Graduate School of Medicine, Japan.,Department of Urology, Chiba University Graduate School of Medicine, Japan
| | - Keiko Mizuno
- Department of Pulmonary Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Japan
| | - Shinichi Sakamoto
- Department of Urology, Chiba University Graduate School of Medicine, Japan
| | - Naohiko Seki
- Department of Functional Genomics, Chiba University Graduate School of Medicine, Japan
| |
Collapse
|
57
|
Lothion-Roy J, Haigh DB, Harris AE, Metzler VM, Alsaleem M, Toss MS, Kariri Y, Ntekim A, Robinson BD, Khani F, Gudas LJ, Allegrucci C, James VH, Madhusudan S, Mather M, Emes RD, Archer N, Fray RG, Rakha E, Jeyapalan JN, Rutland CS, Mongan NP, Woodcock CL. Clinical and molecular significance of the RNA m 6A methyltransferase complex in prostate cancer. Front Genet 2023; 13:1096071. [PMID: 36733939 PMCID: PMC9887525 DOI: 10.3389/fgene.2022.1096071] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/29/2022] [Indexed: 01/13/2023] Open
Abstract
N6-methyladenosine (m6A) is the most abundant internal mRNA modification and is dynamically regulated through distinct protein complexes that methylate, demethylate, and/or interpret the m6A modification. These proteins, and the m6A modification, are involved in the regulation of gene expression, RNA stability, splicing and translation. Given its role in these crucial processes, m6A has been implicated in many diseases, including in cancer development and progression. Prostate cancer (PCa) is the most commonly diagnosed non-cutaneous cancer in men and recent studies support a role for m6A in PCa. Despite this, the literature currently lacks an integrated analysis of the expression of key components of the m6A RNA methyltransferase complex, both in PCa patients and in well-established cell line models. For this reason, this study used immunohistochemistry and functional studies to investigate the mechanistic and clinical significance of the METTL3, METTL14, WTAP and CBLL1 components of the m6A methyltransferase complex in PCa specimens and cell lines. Expression of METTL3 and CBLL1, but not METTL14 and WTAP, was associated with poorer PCa patient outcomes. Expression of METTL3, METTL14, WTAP and CBLL1 was higher in PCa cells compared with non-malignant prostate cells, with the highest expression seen in castrate-sensitive, androgen-responsive PCa cells. Moreover, in PCa cell lines, expression of METTL3 and WTAP was found to be androgen-regulated. To investigate the mechanistic role(s) of the m6A methyltransferase complex in PCa cells, short hairpin RNA (shRNA)-mediated knockdown coupled with next generation sequencing was used to determine the transcriptome-wide roles of METTL3, the catalytic subunit of the m6A methyltransferase complex. Functional depletion of METTL3 resulted in upregulation of the androgen receptor (AR), together with 134 AR-regulated genes. METTL3 knockdown also resulted in altered splicing, and enrichment of cell cycle, DNA repair and metabolic pathways. Collectively, this study identified the functional and clinical significance of four essential m6A complex components in PCa patient specimens and cell lines for the first time. Further studies are now warranted to determine the potential therapeutic relevance of METTL3 inhibitors in development to treat leukaemia to benefit patients with PCa.
Collapse
Affiliation(s)
- Jennifer Lothion-Roy
- Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom,School of Veterinary Medicine and Sciences, University of Nottingham, Sutton Bonington Campus, Loughborough, United Kingdom
| | - Daisy B. Haigh
- Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom,School of Veterinary Medicine and Sciences, University of Nottingham, Sutton Bonington Campus, Loughborough, United Kingdom
| | - Anna E. Harris
- Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom,School of Veterinary Medicine and Sciences, University of Nottingham, Sutton Bonington Campus, Loughborough, United Kingdom
| | - Veronika M. Metzler
- School of Veterinary Medicine and Sciences, University of Nottingham, Sutton Bonington Campus, Loughborough, United Kingdom
| | - Mansour Alsaleem
- Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom,School of Medicine, University of Nottingham, Nottingham, United Kingdom,Department of Applied Medical Science, Applied College, Qassim University, Qassim, Saudi Arabia
| | - Michael S. Toss
- Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom,School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Yousif Kariri
- Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom,School of Medicine, University of Nottingham, Nottingham, United Kingdom,Department of Clinical Laboratory Science, Faculty of Applied Medical Science, Shaqra University, Shaqra, Saudi Arabia
| | - Atara Ntekim
- School of Veterinary Medicine and Sciences, University of Nottingham, Sutton Bonington Campus, Loughborough, United Kingdom,Department of Radiation Oncology, University Hospital Ibadan, University of Ibadan, Ibadan, Nigeria
| | - Brian D. Robinson
- Department of Pathology, Weill Cornell Medicine, New York, NY, United States
| | - Francesca Khani
- Department of Pathology, Weill Cornell Medicine, New York, NY, United States
| | - Lorraine J. Gudas
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States
| | - Cinzia Allegrucci
- Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom,School of Veterinary Medicine and Sciences, University of Nottingham, Sutton Bonington Campus, Loughborough, United Kingdom
| | - Victoria H. James
- Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom,School of Veterinary Medicine and Sciences, University of Nottingham, Sutton Bonington Campus, Loughborough, United Kingdom
| | - Srinivasan Madhusudan
- Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom,School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Melissa Mather
- Faculty of Engineering, University of Nottingham, Nottingham, United Kingdom
| | - Richard D. Emes
- School of Veterinary Medicine and Sciences, University of Nottingham, Sutton Bonington Campus, Loughborough, United Kingdom
| | - Nathan Archer
- School of Veterinary Medicine and Sciences, University of Nottingham, Sutton Bonington Campus, Loughborough, United Kingdom
| | - Rupert G. Fray
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, United Kingdom
| | - Emad Rakha
- School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Jennie N. Jeyapalan
- Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom,School of Veterinary Medicine and Sciences, University of Nottingham, Sutton Bonington Campus, Loughborough, United Kingdom
| | - Catrin S. Rutland
- School of Veterinary Medicine and Sciences, University of Nottingham, Sutton Bonington Campus, Loughborough, United Kingdom
| | - Nigel P. Mongan
- Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom,School of Veterinary Medicine and Sciences, University of Nottingham, Sutton Bonington Campus, Loughborough, United Kingdom,Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States,*Correspondence: Nigel P. Mongan, , ; Corinne L. Woodcock,
| | - Corinne L. Woodcock
- Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom,School of Veterinary Medicine and Sciences, University of Nottingham, Sutton Bonington Campus, Loughborough, United Kingdom,*Correspondence: Nigel P. Mongan, , ; Corinne L. Woodcock,
| |
Collapse
|
58
|
Sreekumar A, Saini S. Role of transcription factors and chromatin modifiers in driving lineage reprogramming in treatment-induced neuroendocrine prostate cancer. Front Cell Dev Biol 2023; 11:1075707. [PMID: 36711033 PMCID: PMC9879360 DOI: 10.3389/fcell.2023.1075707] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 01/02/2023] [Indexed: 01/14/2023] Open
Abstract
Therapy-induced neuroendocrine prostate cancer (NEPC) is a highly lethal variant of prostate cancer that is increasing in incidence with the increased use of next-generation of androgen receptor (AR) pathway inhibitors. It arises via a reversible trans-differentiation process, referred to as neuroendocrine differentiation (NED), wherein prostate cancer cells show decreased expression of AR and increased expression of neuroendocrine (NE) lineage markers including enolase 2 (ENO2), chromogranin A (CHGA) and synaptophysin (SYP). NEPC is associated with poor survival rates as these tumors are aggressive and often metastasize to soft tissues such as liver, lung and central nervous system despite low serum PSA levels relative to disease burden. It has been recognized that therapy-induced NED involves a series of genetic and epigenetic alterations that act in a highly concerted manner in orchestrating lineage switching. In the recent years, we have seen a spurt in research in this area that has implicated a host of transcription factors and epigenetic modifiers that play a role in driving this lineage switching. In this article, we review the role of important transcription factors and chromatin modifiers that are instrumental in lineage reprogramming of prostate adenocarcinomas to NEPC under the selective pressure of various AR-targeted therapies. With an increased understanding of the temporal and spatial interplay of transcription factors and chromatin modifiers and their associated gene expression programs in NEPC, better therapeutic strategies are being tested for targeting NEPC effectively.
Collapse
|
59
|
Malik J, Ahmed S, Momin SS, Shaikh S, Alafnan A, Alanazi J, Said Almermesh MH, Anwar S. Drug Repurposing: A New Hope in Drug Discovery for Prostate Cancer. ACS OMEGA 2023; 8:56-73. [PMID: 36643505 PMCID: PMC9835086 DOI: 10.1021/acsomega.2c05821] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/24/2022] [Indexed: 06/12/2023]
Abstract
Prostate cancer (PCA), the most common cancer in men, accounted for 1.3 million new incidences in 2018. An increase in incidences is an issue of concern that should be addressed. Of all the reported prostate cancers, 85% were detected in stages III and IV, making them difficult to treat. Conventional drugs gradually lose their efficacy due to the developed resistance against them, thus requiring newer therapeutic agents to be used as monotherapy or combination. Recent research regarding treatment options has attained remarkable speed and development. Therefore, in this context, drug repurposing comes into the picture, which is defined as the "investigation of the off-patent, approved and marketed drugs for a novel therapeutic indication" which saves at least 30% of the time and cost, reducing the cost of treatment for patients, which usually runs high in cancer patients. The anticancer property of cardiac glycosides in cancers was tested in the early 1980s. The trend then shifts toward treating prostate cancer by repurposing other cardiovascular drugs. The current review mainly emphasizes the advantageous antiprostate cancer profile of conventional CVS drugs like cardiac glycosides, RAAS inhibitors, statins, heparin, and beta-blockers with underlying mechanisms.
Collapse
Affiliation(s)
- Jonaid
Ahmad Malik
- Department
of Pharmacology and Toxicology, National
Institute of Pharmaceutical Education and Research, Guwahati 781003, India
- Biomedical
Engineering, Indian Institute of Technology
(IIT), Ropar, Punjab 140001, India
| | - Sakeel Ahmed
- Department
of Pharmacology and Toxicology, National
Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat 382355, India
| | - Sadiya Sikandar Momin
- Department
of Pharmaceutics, Annasaheb Dange College of B. Pharmacy, Ashta, Shivaji University, Sangli, Maharastra 416301, India
| | - Sijal Shaikh
- Sandip Institute
of Pharmaceutical Sciences, Savitribai Phule
Pune University, Nashik, Maharashtra 422213, India
| | - Ahmed Alafnan
- Department
of Pharmacology and Toxicology, University
of Hail, Hail 81422, Saudi Arabia
| | - Jowaher Alanazi
- Department
of Pharmacology and Toxicology, University
of Hail, Hail 81422, Saudi Arabia
| | | | - Sirajudheen Anwar
- Department
of Pharmacology and Toxicology, University
of Hail, Hail 81422, Saudi Arabia
| |
Collapse
|
60
|
Matias-Barrios VM, Dong X. The Implication of Topoisomerase II Inhibitors in Synthetic Lethality for Cancer Therapy. Pharmaceuticals (Basel) 2023; 16:94. [PMID: 36678591 PMCID: PMC9866718 DOI: 10.3390/ph16010094] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/31/2022] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
DNA topoisomerase II (Top2) is essential for all eukaryotic cells in the regulation of DNA topology through the generation of temporary double-strand breaks. Cancer cells acquire enhanced Top2 functions to cope with the stress generated by transcription and DNA replication during rapid cell division since cancer driver genes such as Myc and EZH2 hijack Top2 in order to realize their oncogenic transcriptomes for cell growth and tumor progression. Inhibitors of Top2 are therefore designed to target Top2 to trap it on DNA, subsequently causing protein-linked DNA breaks, a halt to the cell cycle, and ultimately cell death. Despite the effectiveness of these inhibitors, cancer cells can develop resistance to them, thereby limiting their therapeutic utility. To maximize the therapeutic potential of Top2 inhibitors, combination therapies to co-target Top2 with DNA damage repair (DDR) machinery and oncogenic pathways have been proposed to induce synthetic lethality for more thorough tumor suppression. In this review, we will discuss the mode of action of Top2 inhibitors and their potential applications in cancer treatments.
Collapse
Affiliation(s)
- Victor M. Matias-Barrios
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada
- School of Medicine and Health Sciences, Tecnologico de Monterrey, Avenida Eugenio Garza Sada 2501, Monterrey 64849, Mexico
| | - Xuesen Dong
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada
| |
Collapse
|
61
|
Xu C, Zhao S, Cai L. Epigenetic (De)regulation in Prostate Cancer. Cancer Treat Res 2023; 190:321-360. [PMID: 38113006 PMCID: PMC11421856 DOI: 10.1007/978-3-031-45654-1_10] [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: 12/21/2023]
Abstract
Prostate cancer (PCa) is a heterogeneous disease exhibiting both genetic and epigenetic deregulations. Epigenetic alterations are defined as changes not based on DNA sequence, which include those of DNA methylation, histone modification, and chromatin remodeling. Androgen receptor (AR) is the main driver for PCa and androgen deprivation therapy (ADT) remains a backbone treatment for patients with PCa; however, ADT resistance almost inevitably occurs and advanced diseases develop termed castration-resistant PCa (CRPC), due to both genetic and epigenetic changes. Due to the reversible nature of epigenetic modifications, inhibitors targeting epigenetic factors have become promising anti-cancer agents. In this chapter, we focus on recent studies about the dysregulation of epigenetic regulators crucially involved in the initiation, development, and progression of PCa and discuss the potential use of inhibitors targeting epigenetic modifiers for treatment of advanced PCa.
Collapse
Affiliation(s)
- Chenxi Xu
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Shuai Zhao
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Ling Cai
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA.
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA.
| |
Collapse
|
62
|
Pituitary-Gland-Based Genes Participates in Intrauterine Growth Restriction in Piglets. Genes (Basel) 2022; 13:genes13112141. [DOI: 10.3390/genes13112141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
Intrauterine growth restriction (IUGR) is a major problem associated with piglet growth performance. The incidence of IUGR is widespread in Rongchang pigs. The pituitary gland is important for regulating growth and metabolism, and research has identified genes associated with growth and development. The pituitary gland of newborn piglets with normal birth weight (NBW group, n = 3) and (IUGR group, n = 3) was collected for transcriptome analysis. A total of 323 differentially expression genes (DEGs) were identified (|log2(fold-change)| > 1 and q value < 0.05), of which 223 were upregulated and 100 were downregulated. Gene Ontology (GO) functional and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses showed that the DEGs were mainly related to the extracellular matrix, regulation of the multicellular organismal process, tissue development and angiogenesis, which participate in the growth and immune response in IUGR piglets. Moreover, 7 DEGs including IGF2, THBS1, ITGA1, ITGA8, EPSTI1, FOSB, and UCP2 were associated with growth and immune response. Furthermore, based on the interaction network analysis of the DEGs, two genes, IGF2 and THBS1, participated in cell proliferation, embryonic development and angiogenesis. IGF2 and THBS1 were also the main genes participating in the IUGR. This study identified the core genes involved in IUGR in piglets and provided a reference for exploring the effect of the pituitary gland on piglet growth.
Collapse
|
63
|
Liu S, Alabi BR, Yin Q, Stoyanova T. Molecular mechanisms underlying the development of neuroendocrine prostate cancer. Semin Cancer Biol 2022; 86:57-68. [PMID: 35597438 DOI: 10.1016/j.semcancer.2022.05.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/19/2022] [Accepted: 05/14/2022] [Indexed: 01/27/2023]
Abstract
Prostate cancer is the most common non-cutaneous cancer and the second leading cause of cancer-associated deaths among men in the United States. Androgen deprivation therapy (ADT) is the standard of care for advanced prostate cancer. While patients with advanced prostate cancer initially respond to ADT, the disease frequently progresses to a lethal metastatic form, defined as castration-resistant prostate cancer (CRPC). After multiple rounds of anti-androgen therapies, 20-25% of metastatic CRPCs develop a neuroendocrine (NE) phenotype. These tumors are classified as neuroendocrine prostate cancer (NEPC). De novo NEPC is rare and accounts for less than 2% of all prostate cancers at diagnosis. NEPC is commonly characterized by the expression of NE markers and the absence of androgen receptor (AR) expression. NEPC is usually associated with tumor aggressiveness, hormone therapy resistance, and poor clinical outcome. Here, we review the molecular mechanisms underlying the emergence of NEPC and provide insights into the future perspectives on potential therapeutic strategies for NEPC.
Collapse
Affiliation(s)
- Shiqin Liu
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Palo Alto, CA, USA
| | - Busola Ruth Alabi
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Palo Alto, CA, USA
| | - Qingqing Yin
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Palo Alto, CA, USA
| | - Tanya Stoyanova
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Palo Alto, CA, USA.
| |
Collapse
|
64
|
Asberry AM, Cai X, Deng X, Liu S, Santiago U, Sims H, Liang W, Xu X, Wan J, Jiang W, Camacho C, Dai M, Hu CD. Discovery and Biological Characterization of PRMT5:MEP50 Protein-Protein Interaction Inhibitors. J Med Chem 2022; 65:13793-13812. [PMID: 36206451 PMCID: PMC11167723 DOI: 10.1021/acs.jmedchem.2c01000] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Protein arginine methyltransferase 5 (PRMT5) is a master epigenetic regulator and an extensively validated therapeutic target in multiple cancers. Notably, PRMT5 is the only PRMT that requires an obligate cofactor, methylosome protein 50 (MEP50), to function. We developed compound 17, a novel small-molecule PRMT5:MEP50 protein-protein interaction (PPI) inhibitor, after initial virtual screen hit identification and analogue refinement. Molecular docking indicated that compound 17 targets PRMT5:MEP50 PPI by displacing the MEP50 W54 burial into a hydrophobic pocket of the PRMT5 TIM barrel. In vitro analysis indicates IC50 < 500 nM for prostate and lung cancer cells with selective, specific inhibition of PRMT5:MEP50 substrate methylation and target gene expression, and RNA-seq analysis suggests that compound 17 may dysregulate TGF-β signaling. Compound 17 provides a proof of concept in targeting PRMT5:MEP50 PPI, as opposed to catalytic targeting, as a novel mechanism of action and supports further preclinical development of inhibitors in this class.
Collapse
Affiliation(s)
- Andrew M. Asberry
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA
- Purdue University Interdisciplinary Life Sciences Graduate Program, Purdue University, West Lafayette, IN, 47907, USA
| | - Xinpei Cai
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907, USA
| | - Xuehong Deng
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- The Indiana University Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, IN, 46202, USA
| | - Ulises Santiago
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Hunter Sims
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907, USA
| | - Weida Liang
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907, USA
| | - Xueyong Xu
- Department of Biological Sciences, Purdue University, 240 S Martin Jischke Drive, West Lafayette, IN 47907, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- The Indiana University Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, IN, 46202, USA
- The Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Wen Jiang
- Department of Biological Sciences, Purdue University, 240 S Martin Jischke Drive, West Lafayette, IN 47907, USA
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Carlos Camacho
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Mingji Dai
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907, USA
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Chang-Deng Hu
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA
| |
Collapse
|
65
|
Wang Y, Wu N, Wang K, Liao Y, Guo J, Zhong B, Guo T, Liang J, Jiang N. Specific classification and new therapeutic targets for neuroendocrine prostate cancer: A patient-based, diagnostic study. Front Genet 2022; 13:955133. [PMID: 36118857 PMCID: PMC9479159 DOI: 10.3389/fgene.2022.955133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: Neuroendocrine prostate cancer (NEPC) is an aggressive variant of prostate cancer (PC) that may arise de novo or in patients previously treated with hormonal therapies for prostate adenocarcinoma as a mechanism of resistance. In our investigation, there appeared to be a strong correlation between neuroendocrine differentiation prostate cancer (NEDPC) and NEPC. The objectives of this study included exploring whether NEDPC is an intermediate stage in the progression of high-risk prostate cancer (HRPC) to NEPC and identifying risk factors and new targets associated with survival in the treatment of NEPC. Methods: The selected prostate cancer patients were progressed to high-risk and characterized by neuroendocrine. We collected the clinical data and characteristics of patients with three types of cancer: the incidence of metastasis, site and time of metastasis, recurrence rate, related treatment methods, etc. The similarity and differences of the three groups were compared through experiment and database. Results: By analyzing the clinical data and immunohistochemical results, we found that there seems to be a clinical feature of neuroendocrine differentiation (NED) status in between when patients progress from PC to NEPC. Finding novel treatment targets would therefore be beneficial by taking into account NEDPC as the stage of PC progression prior to NEPC. The metastasis-free survival curve and the immunohistochemical results are informing us that NEDPC can be a pre-state for diagnosing NEPC. Conclusion: NEPC is a late PC symptom that is frequently disregarded and has a bad prognosis. Finding novel treatment targets would therefore be beneficial by taking into account NEDPC as the stage of PC progression prior to NEPC.
Collapse
Affiliation(s)
- YouZhi Wang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Ning Wu
- Key Laboratory of Breast Cancer Prevention and Therapy, State Ministry of Education, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Clinical Research Center for Cancer, Tianjin Medical University Cancer Hospital and Institute, Tianjin, China
| | - KeKe Wang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - YiHao Liao
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - JiaNing Guo
- Department of Pathology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - BoQiang Zhong
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Tao Guo
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - JiaMing Liang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Ning Jiang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
66
|
Yabusaki R, Yoshimura K, Taku K, Suzuki M. Prostate cancer recurring as small‐cell carcinoma with a
BRCA2
somatic mutation. IJU Case Rep 2022; 5:489-492. [PMID: 36341199 PMCID: PMC9626344 DOI: 10.1002/iju5.12523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 07/26/2022] [Indexed: 11/25/2022] Open
Abstract
Introduction Small‐cell carcinoma of the prostate has a poor prognosis, and treatment options for the refractory disease are unclear. Case presentation A 68‐year‐old man with prostate cancer was referred to our hospital. He was treated with combined androgen blockade (bicalutamide and degarelix acetate). The disease progressed to castration‐resistant prostate cancer, but with additional treatment, prostate‐specific antigen levels remained below 0.02 ng/mL. However, computed tomography revealed enlarged right inguinal lymph nodes; moreover, his neuron‐specific enolase levels were elevated. Histopathologic analysis of a biopsied lymph node confirmed small‐cell carcinoma. After administering cytotoxic chemotherapy (etoposide plus cisplatin and amrubicin), the patient temporarily improved before relapsing. After genetic testing of the biopsy specimen revealed a BRCA2 deletion, we administered the oral PARP‐2 inhibitor olaparib, which has achieved partial remission for 8 months. Conclusion PARP‐2 inhibition may improve the survival of patients with BRCA2‐positive small‐cell carcinoma of the prostate.
Collapse
Affiliation(s)
- Ryo Yabusaki
- Department of Urology Shizuoka General Hospital Shizuoka Japan
| | - Koji Yoshimura
- Department of Urology Shizuoka General Hospital Shizuoka Japan
| | - Keisei Taku
- Department of Oncology Shizuoka General Hospital Shizuoka Japan
| | - Makoto Suzuki
- Department of Pathology Shizuoka General Hospital Shizuoka Japan
| |
Collapse
|
67
|
Chen L, Yu C, Xiong Y, Chen K, Liu P, Panayi AC, Xiao X, Feng Q, Mi B, Liu G. Multifunctional hydrogel enhances bone regeneration through sustained release of Stromal Cell-Derived Factor-1α and exosomes. Bioact Mater 2022; 25:460-471. [PMID: 37056272 PMCID: PMC10087917 DOI: 10.1016/j.bioactmat.2022.07.030] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/24/2022] [Accepted: 07/27/2022] [Indexed: 11/02/2022] Open
Abstract
Fracture nonunion remains a great challenge for orthopedic surgeons. Fracture repair comprises of three phases, the inflammatory, repair and remodeling stage. Extensive advancements have been made in the field of bone repair, including development of strategies to balance the M1/M2 macrophage populations, and to improve osteogenesis and angiogenesis. However, such developments focused on only one or the latter two phases, while ignoring the inflammatory phase during which cell recruitment occurs. In this study, we combined Stromal Cell-Derived Factor-1α (SDF-1α) and M2 macrophage derived exosomes (M2D-Exos) with a hyaluronic acid (HA)-based hydrogel precursor solution to synthesize an injectable, self-healing, adhesive HA@SDF-1α/M2D-Exos hydrogel. The HA hydrogel demonstrated good biocompatibility and hemostatic ability, with the 4% HA hydrogels displaying great antibacterial activity against gram-negative E. coli and gram-positive S. aureus and Methicillin-resistant Staphylococcus aureus (MRSA). Synchronously and sustainably released SDF-1α and M2D-Exos from the HA@SDF-1α/M2D-Exos hydrogel enhanced proliferation and migration of human bone marrow mesenchymal stem cell (HMSCs) and Human Umbilical Vein Endothelial Cells (HUVECs), promoting osteogenesis and angiogenesis both in vivo and in vitro. Overall, the developed HA@ SDF-1α/M2D-Exos hydrogel was compatible with the natural healing process of fractures and provides a new modality for accelerating bone repair by coupling osteogenesis, angiogenesis, and resisting infection at all stages.
Collapse
|
68
|
Zhou H, He Q, Li C, Alsharafi BLM, Deng L, Long Z, Gan Y. Focus on the tumor microenvironment: A seedbed for neuroendocrine prostate cancer. Front Cell Dev Biol 2022; 10:955669. [PMID: 35938167 PMCID: PMC9355504 DOI: 10.3389/fcell.2022.955669] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
The tumor microenvironment (TME) is a microecology consisting of tumor and mesenchymal cells and extracellular matrices. The TME plays important regulatory roles in tumor proliferation, invasion, metastasis, and differentiation. Neuroendocrine differentiation (NED) is a mechanism by which castration resistance develops in advanced prostate cancer (PCa). NED is induced after androgen deprivation therapy and neuroendocrine prostate cancer (NEPC) is established finally. NEPC has poor prognosis and short overall survival and is a major cause of death in patients with PCa. Both the cellular and non-cellular components of the TME regulate and induce NEPC formation through various pathways. Insights into the roles of the TME in NEPC evolution, growth, and progression have increased over the past few years. These novel insights will help refine the NEPC formation model and lay the foundation for the discovery of new NEPC therapies targeting the TME.
Collapse
Affiliation(s)
- Hengfeng Zhou
- Andrology Center, Department of Urology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Qiangrong He
- Andrology Center, Department of Urology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Chao Li
- Andrology Center, Department of Urology, the Third Xiangya Hospital, Central South University, Changsha, China
| | | | - Liang Deng
- Andrology Center, Department of Urology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhi Long
- Andrology Center, Department of Urology, the Third Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Zhi Long, ; Yu Gan,
| | - Yu Gan
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Zhi Long, ; Yu Gan,
| |
Collapse
|
69
|
Islam R, Mishra J, Polavaram NS, Bhattacharya S, Hong Z, Bodas S, Sharma S, Bouska A, Gilbreath T, Said AM, Smith LM, Teply BA, Muders MH, Batra SK, Datta K, Dutta S. Neuropilin-2 axis in regulating secretory phenotype of neuroendocrine-like prostate cancer cells and its implication in therapy resistance. Cell Rep 2022; 40:111097. [PMID: 35858551 PMCID: PMC9362995 DOI: 10.1016/j.celrep.2022.111097] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 04/06/2022] [Accepted: 06/23/2022] [Indexed: 12/25/2022] Open
Abstract
Neuroendocrine (NE)-like tumors secrete various signaling molecules to establish paracrine communication within the tumor milieu and to create a therapy-resistant environment. It is important to identify molecular mediators that regulate this secretory phenotype in NE-like cancer. The current study highlights the importance of a cell surface molecule, Neuropilin-2 (NRP2), for the secretory function of NE-like prostate cancer (PCa). Our analysis on different patient cohorts suggests that NRP2 is high in NE-like PCa. We have developed cell line models to investigate NRP2's role in NE-like PCa. Our bioinformatics, mass spectrometry, cytokine array, and other supporting experiments reveal that NRP2 regulates robust secretory phenotype in NE-like PCa and controls the secretion of factors promoting cancer cell survival. Depletion of NRP2 reduces the secretion of these factors and makes resistant cancer cells sensitive to chemotherapy in vitro and in vivo. Therefore, targeting NRP2 can revert cellular secretion and sensitize PCa cells toward therapy.
Collapse
Affiliation(s)
- Ridwan Islam
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, BCC, Omaha, NE 68198, USA
| | - Juhi Mishra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, BCC, Omaha, NE 68198, USA
| | - Navatha Shree Polavaram
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, BCC, Omaha, NE 68198, USA
| | - Sreyashi Bhattacharya
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, BCC, Omaha, NE 68198, USA
| | - Zhengdong Hong
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, BCC, Omaha, NE 68198, USA
| | - Sanika Bodas
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, BCC, Omaha, NE 68198, USA
| | - Sunandini Sharma
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, BCC, Omaha, NE 68198, USA
| | - Alyssa Bouska
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, BCC, Omaha, NE 68198, USA
| | - Tyler Gilbreath
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, BCC, Omaha, NE 68198, USA
| | - Ahmed M Said
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Helwan University, Ein-Helwan, Helwan, Cairo, Egypt
| | - Lynette M Smith
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, BCC, Omaha, NE 68198, USA
| | - Benjamin A Teply
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, BCC, Omaha, NE 68198, USA
| | - Michael H Muders
- Department of Prostate Cancer Research, Center for Pathology, University of Bonn Medical Center, Bonn, Germany
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, BCC, Omaha, NE 68198, USA
| | - Kaustubh Datta
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, BCC, Omaha, NE 68198, USA.
| | - Samikshan Dutta
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, BCC, Omaha, NE 68198, USA.
| |
Collapse
|
70
|
Integrative Analysis of Bulk RNA-Seq and Single-Cell RNA-Seq Unveils the Characteristics of the Immune Microenvironment and Prognosis Signature in Prostate Cancer. JOURNAL OF ONCOLOGY 2022; 2022:6768139. [PMID: 35909899 PMCID: PMC9325591 DOI: 10.1155/2022/6768139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/10/2022] [Accepted: 06/21/2022] [Indexed: 12/01/2022]
Abstract
The immune microenvironment is a culmination of the collaborative effort of immune cells and is important in cancer development. The underlying mechanisms of the tumor immune microenvironment in regulating prostate cancer (PRAD) are unclear. In the current study, 144 natural killer cell-related genes were identified using differential expression, single-sample gene set enrichment analysis, and weighted gene coexpression network analysis. Furthermore, VCL, ACTA2, MYL9, MYLK, MYH11, TPM1, ACTG2, TAGLN, and FLNC were selected as hub genes via the protein-protein interaction network. Based on the expression patterns of the hub genes, endothelial, epithelial, and tissue stem cells were identified as key cell subpopulations, which could regulate PRAD via immune response, extracellular signaling, and protein formation. Moreover, 27 genes were identified as prognostic signatures and used to construct the risk score model. Receiver operating characteristic curves revealed the good performance of the risk score model in both the training and testing datasets. Different chemotherapeutic responses were observed between the low- and high-risk groups. Additionally, a nomogram based on the risk score and other clinical features was established to predict the 1-, 3-, and 5-year progression-free interval of patients with PRAD. This study provides novel insights into the molecular mechanisms of the immune microenvironment and its role in the pathogenesis of PARD. The identification of key cell subpopulations has a potential therapeutic and prognostic use in PRAD.
Collapse
|
71
|
Gu J, Zhu N, Li HF, Zhang CJ, Gong YZ, Liao DF, Qin L. Ezetimibe and Cancer: Is There a Connection? Front Pharmacol 2022; 13:831657. [PMID: 35924044 PMCID: PMC9340271 DOI: 10.3389/fphar.2022.831657] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 06/23/2022] [Indexed: 11/29/2022] Open
Abstract
The high level of serum cholesterol caused by the excessive absorption of cholesterol can lead to hypercholesteremia, thus promoting the occurrence and development of cancer. Ezetimibe is a drug that reduces cholesterol absorption and has been widely used for the treatment of patients with high circulating cholesterol levels for many years. Mechanistically, ezetimibe works by binding to NPC1L1, which is a key mediator of cholesterol absorption. Accumulating data from preclinical models have shown that ezetimibe alone could inhibit the development and progression of cancer through a variety of mechanisms, including anti-angiogenesis, stem cell suppression, anti-inflammation, immune enhancement and anti-proliferation. In the past decade, there has been heated discussion on whether ezetimibe combined with statins will increase the risk of cancer. At present, more and more evidence shows that ezetimibe does not increase the risk of cancers, which supports the role of ezetimibe in anti-cancer. In this review, we discussed the latest progress in the anti-cancer properties of ezetimibe and elucidated its underlying molecular mechanisms. Finally, we highlighted the potential of ezetimibe as a therapeutic agent in future cancer treatment and prevention.
Collapse
Affiliation(s)
- Jia Gu
- Laboratory of Stem Cell Regulation With Chinese Medicine and its Application, HunanUniversity of Chinese Medicine, Changsha, China
| | - Neng Zhu
- Department of Urology, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Hong-Fang Li
- Laboratory of Stem Cell Regulation With Chinese Medicine and its Application, HunanUniversity of Chinese Medicine, Changsha, China
| | - Chan-Juan Zhang
- Laboratory of Stem Cell Regulation With Chinese Medicine and its Application, HunanUniversity of Chinese Medicine, Changsha, China
| | - Yong-Zhen Gong
- Laboratory of Stem Cell Regulation With Chinese Medicine and its Application, HunanUniversity of Chinese Medicine, Changsha, China
| | - Duan-Fang Liao
- Laboratory of Stem Cell Regulation With Chinese Medicine and its Application, HunanUniversity of Chinese Medicine, Changsha, China
| | - Li Qin
- Laboratory of Stem Cell Regulation With Chinese Medicine and its Application, HunanUniversity of Chinese Medicine, Changsha, China
- Institutional Key Laboratory of Vascular Biology and Translational Medicine in Hunan Province, Changsha, China
- Hunan Province Engineering Research Center of Bioactive Substance Discovery of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- *Correspondence: Li Qin,
| |
Collapse
|
72
|
Shi Y, Li J, Chen H, Hu Y, Tang L, Wang Y, Zang X, Ma X, Huang G, Zhou X, Tao M, lv Z, Chen S, Qiu A, Zhuang S, Liu N. Inhibition of EZH2 suppresses peritoneal angiogenesis by targeting a VEGFR2/ERK1/2/HIF‐1α dependent signaling pathway. J Pathol 2022; 258:164-178. [PMID: 35792675 DOI: 10.1002/path.5987] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/10/2022] [Accepted: 07/04/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Yingfeng Shi
- Department of Nephrology, Shanghai East Hospital Tongji University School of Medicine Shanghai PR China
| | - Jinqing Li
- Department of Nephrology, Shanghai East Hospital Tongji University School of Medicine Shanghai PR China
| | - Hui Chen
- Department of Nephrology, Shanghai East Hospital Tongji University School of Medicine Shanghai PR China
| | - Yan Hu
- Department of Nephrology, Shanghai East Hospital Tongji University School of Medicine Shanghai PR China
| | - Lunxian Tang
- Emergency department of critical care medicine, Shanghai East Hospital Tongji University School of Medicine Shanghai PR China
| | - Yi Wang
- Department of Nephrology, Shanghai East Hospital Tongji University School of Medicine Shanghai PR China
| | - Xiujuan Zang
- Department of Nephrology Shanghai Songjiang District Central Hospital Shanghai PR China
| | - Xiaoyan Ma
- Department of Nephrology, Shanghai East Hospital Tongji University School of Medicine Shanghai PR China
| | - Guansen Huang
- Department of Nephrology, Shanghai East Hospital Tongji University School of Medicine Shanghai PR China
| | - Xun Zhou
- Department of Nephrology, Shanghai East Hospital Tongji University School of Medicine Shanghai PR China
| | - Min Tao
- Department of Nephrology, Shanghai East Hospital Tongji University School of Medicine Shanghai PR China
| | - Zexin lv
- Department of Nephrology, Shanghai East Hospital Tongji University School of Medicine Shanghai PR China
| | - Si Chen
- Department of Nephrology, Shanghai East Hospital Tongji University School of Medicine Shanghai PR China
| | - Andong Qiu
- School of Life Science and Technology, Advanced Institute of Translational Medicine Tongji University Shanghai PR China
| | - Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital Tongji University School of Medicine Shanghai PR China
- Department of Medicine, Rhode Island Hospital and Alpert Medical School Brown University Providence RI USA
| | - Na Liu
- Department of Nephrology, Shanghai East Hospital Tongji University School of Medicine Shanghai PR China
| |
Collapse
|
73
|
Bremer SCB, Bittner G, Elakad O, Dinter H, Gaedcke J, König AO, Amanzada A, Ellenrieder V, Freiherr von Hammerstein-Equord A, Ströbel P, Bohnenberger H. Enhancer of Zeste Homolog 2 (EZH2) Is a Marker of High-Grade Neuroendocrine Neoplasia in Gastroenteropancreatic and Pulmonary Tract and Predicts Poor Prognosis. Cancers (Basel) 2022; 14:cancers14122828. [PMID: 35740494 PMCID: PMC9221317 DOI: 10.3390/cancers14122828] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/29/2022] [Accepted: 06/04/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Neuroendocrine neoplasms most frequently arise in the gastroenteropancreatic and pulmonary tract and show an increasing incidence and prevalence. The prognosis and treatment depend on tumor proliferation and clinical behavior. Highly proliferating grade 3 neoplasms especially, show a wildly divergent therapy response and prognosis. In particular, it is crucial to securely separate the more indolent G3 tumors from the more aggressive carcinomas. Currently, this distinction is based on a combination of clinical, morphologic, immunohistochemical, and molecular biomarkers. However, none of these markers allow for a reliable distinction, and additional markers are needed. EZH2 has attracted increasing interest in different tumor entities. We aimed to analyze the expression of EZH2 in different neuroendocrine neoplasms and to correlate the expression with clinical parameters and survival. We demonstrate that EZH2 is nearly exclusively expressed in highly proliferative neoplasms and is a robust biomarker for identifying aggressive G3 tumors with poor prognosis. Abstract Tumor grading is a robust prognostic predictor in patients with neuroendocrine neoplasms (NEN) and guides therapy, especially in tumors with high proliferation. NEN can be separated into well-differentiated and poorly differentiated types. The more aggressive NEN have been further separated into neuroendocrine tumors (NET G3) with a better prognosis and neuroendocrine carcinomas (NEC) with a worse prognosis. Despite this distinction’s tremendous clinical and therapeutic relevance, optimal diagnostic biomarkers are still lacking. In this study, we analyzed the protein expression and prognostic impact of Enhancer of Zeste Homolog 2 (EZH2) by immunohistochemistry in 219 tissue samples of gastroenteropancreatic (GEP-NEN) and pulmonary NEN (P-NEN). EZH2 was almost exclusively expressed in NEN with a proliferation rate above 20% (G3), while all low-grade tumors were nearly negative. Among high-grade NEN, 65% showed high and 35% low expression of EZH2. In this group, the high expression of EZH2 was significantly associated with poor overall survival and NEC histology. Interestingly, EZH2 seems to act independently of Polycomb Repressive Complex 2 (PRC2) in NEN. In conclusion, we propose EZH2 as a robust biomarker for distinguishing between NET G3 and NEC among gastroenteropancreatic and pulmonary NEN.
Collapse
Affiliation(s)
- Sebastian C. B. Bremer
- Clinic for Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Goettingen, Georg-August-University, 37075 Goettingen, Germany; (A.O.K.); (A.A.); (V.E.)
- Correspondence:
| | - Gabi Bittner
- Institute of Pathology, University Medical Center Goettingen, Georg-August-University, 37075 Goettingen, Germany; (G.B.); (O.E.); (H.D.); (P.S.); (H.B.)
| | - Omar Elakad
- Institute of Pathology, University Medical Center Goettingen, Georg-August-University, 37075 Goettingen, Germany; (G.B.); (O.E.); (H.D.); (P.S.); (H.B.)
| | - Helen Dinter
- Institute of Pathology, University Medical Center Goettingen, Georg-August-University, 37075 Goettingen, Germany; (G.B.); (O.E.); (H.D.); (P.S.); (H.B.)
| | - Jochen Gaedcke
- Clinic for General, Visceral and Pediatric Surgery, University Medical Center Goettingen, Georg-August-University, 37075 Goettingen, Germany;
| | - Alexander O. König
- Clinic for Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Goettingen, Georg-August-University, 37075 Goettingen, Germany; (A.O.K.); (A.A.); (V.E.)
| | - Ahmad Amanzada
- Clinic for Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Goettingen, Georg-August-University, 37075 Goettingen, Germany; (A.O.K.); (A.A.); (V.E.)
| | - Volker Ellenrieder
- Clinic for Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Goettingen, Georg-August-University, 37075 Goettingen, Germany; (A.O.K.); (A.A.); (V.E.)
| | | | - Philipp Ströbel
- Institute of Pathology, University Medical Center Goettingen, Georg-August-University, 37075 Goettingen, Germany; (G.B.); (O.E.); (H.D.); (P.S.); (H.B.)
| | - Hanibal Bohnenberger
- Institute of Pathology, University Medical Center Goettingen, Georg-August-University, 37075 Goettingen, Germany; (G.B.); (O.E.); (H.D.); (P.S.); (H.B.)
| |
Collapse
|
74
|
Xie Y, Ning S, Hu J. Molecular mechanisms of neuroendocrine differentiation in prostate cancer progression. J Cancer Res Clin Oncol 2022; 148:1813-1823. [PMID: 35633416 PMCID: PMC9189092 DOI: 10.1007/s00432-022-04061-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/09/2022] [Indexed: 12/03/2022]
Abstract
Background Rapid evolution of the therapeutic management of prostate cancer, especially in in second-generation androgen inhibitors, has increased the opportunity of transformation from prostate cancer (PCa) to neuroendocrine prostate cancer (NEPC). NEPC still lacks effective diagnostic and therapeutic interventions. Researches into the molecular characteristics of neuroendocrine differentiation is undoubtedly crucial to the discovery of new target genes for accurate diagnostic and therapeutic targets. Purpose In this review, we focus on the relevant genes and molecular mechanisms that have contributed to the transformation in the progression of PCa and discuss the potential targeted molecule that might improve diagnostic accuracy and therapeutic effectiveness. Methods The relevant literatures from PubMed have been reviewed for this article. Conclusion Several molecular characteristics influence the progression of neuroendocrine differentiation of prostate cancer which will provide a novel sight for accurate diagnosis and target therapeutic intervention for patients with NEPC.
Collapse
Affiliation(s)
- Yuchen Xie
- Affiliated Renmin Hospital of Jiangsu University, Zhenjiang First People's Hospital, Zhenjiang, 212002, China
| | - Songyi Ning
- Jiangsu University, Zhenjiang, 212013, China
| | - Jianpeng Hu
- Affiliated Renmin Hospital of Jiangsu University, Zhenjiang First People's Hospital, Zhenjiang, 212002, China.
| |
Collapse
|
75
|
Zhang R, Ma L, Wei Y, Wei K, Song T, Du Z, Feng Z. KIF22 Promotes Development of Pancreatic Cancer by Regulating the MEK/ERK/P21 Signaling Axis. BIOMED RESEARCH INTERNATIONAL 2022; 2022:6000925. [PMID: 35578724 PMCID: PMC9107036 DOI: 10.1155/2022/6000925] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/28/2022] [Accepted: 04/12/2022] [Indexed: 12/24/2022]
Abstract
The study is aimed at exploring the potential biological process and molecular mechanism of KIF22 involved in the development and progression of pancreatic cancer. First, we used the GEPIA database and tissue qRT-PCR to examine the expression of KIF22 mRNA in pancreatic cancer. Meanwhile, immunohistochemistry revealed the presence of KIF22 in 71 pancreatic cancer tissues versus 30 paracarcinoma tissues. Then, we also explored the relationship between KIF22 expression level and clinical prognosis. Furthermore, in pancreatic cancer cells, we silenced KIF22 by transfecting KIF22 SiRNA, and we investigated the effect of KIF22 on the proliferation of pancreatic cancer cells with MTT and colony formation assays. Finally, we used Gene Set Enrichment Analysis (GSEA) to look at the effect of KIF22 on the cell cycle regulation of pancreatic cancer cells, and we used Western blot to look at the relationship between KIF22 and the phosphorylated MEK1/2, ERK1/2 (p-MEK1/2, p-ERK1/2), and the cyclin-dependent kinase inhibitor (P21). In this study, we found that KIF22 was highly expressed in pancreatic cancer tissues, and patients with high expression of KIF22 demonstrated significantly worse clinical prognosis outcomes (P < 0.05). When the KIF22 gene was silenced in pancreatic cancer cells (PANC-1 and MIA PaCa-2), the cells' ability to proliferate was significantly reduced. Furthermore, GSEA confirmed that KIF22 is involved in cell cycle regulation in pancreatic cancer patients (FDR = 0.00158, P < 0.0001). Besides, the level of KIF22 expression was positively correlated with Ki67 (r = 0.8043, P < 0.0001), and KIF22 can promote the transmutation of G1/S. The expression of p-MEK1/2 and p-ERK1/2 was significantly downregulated, while P21 expression was significantly upregulated (P < 0.05). According to our findings, KIF22 is highly expressed in pancreatic cancer and demonstrates a poor clinical prognosis. It regulates the cell cycle via the MEK/ERK/P21 signaling axis and promotes the development of pancreatic cancer.
Collapse
Affiliation(s)
- Ruiyun Zhang
- Southern University of Science and Technology Hospital, No. 6019, Liuxian Avenue, Nanshan District, Shenzhen, Guangdong, China
| | - Li Ma
- Gansu Provincial Hospital, No. 204, Donggang West Road, Chengguan District, Lanzhou, Gansu, China
| | - Yucai Wei
- Lanzhou University Second Hospital, No. 82, Cuiyingmen, Chengguan District, Lanzhou, Gansu, China
| | - Kongkong Wei
- Lanzhou University Second Hospital, No. 82, Cuiyingmen, Chengguan District, Lanzhou, Gansu, China
| | - Tianliang Song
- Lanzhou University Second Hospital, No. 82, Cuiyingmen, Chengguan District, Lanzhou, Gansu, China
| | - Zhixing Du
- Lanzhou University Second Hospital, No. 82, Cuiyingmen, Chengguan District, Lanzhou, Gansu, China
| | - Zhijun Feng
- Southern University of Science and Technology Hospital, No. 6019, Liuxian Avenue, Nanshan District, Shenzhen, Guangdong, China
| |
Collapse
|
76
|
Huang J, Lin B, Li B. Anti-Androgen Receptor Therapies in Prostate Cancer: A Brief Update and Perspective. Front Oncol 2022; 12:865350. [PMID: 35372068 PMCID: PMC8965587 DOI: 10.3389/fonc.2022.865350] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 02/17/2022] [Indexed: 12/28/2022] Open
Abstract
Prostate cancer is a major health issue in western countries and is the second leading cause of cancer death in American men. Prostate cancer depends on the androgen receptor (AR), a transcriptional factor critical for prostate cancer growth and progression. Castration by surgery or medical treatment reduces androgen levels, resulting in prostatic atrophy and prostate cancer regression. Thus, metastatic prostate cancers are initially managed with androgen deprivation therapy. Unfortunately, prostate cancers rapidly relapse after castration therapy and progress to a disease stage called castration-resistant prostate cancer (CRPC). Currently, clinical treatment for CRPCs is focused on suppressing AR activity with antagonists like Enzalutamide or by reducing androgen production with Abiraterone. In clinical practice, these treatments fail to yield a curative benefit in CRPC patients in part due to AR gene mutations or splicing variations, resulting in AR reactivation. It is conceivable that eliminating the AR protein in prostate cancer cells is a promising solution to provide a potential curative outcome. Multiple strategies have emerged, and several potent agents that reduce AR protein levels were reported to eliminate xenograft tumor growth in preclinical models via distinct mechanisms, including proteasome-mediated degradation, heat-shock protein inhibition, AR splicing suppression, blockage of AR nuclear localization, AR N-terminal suppression. A few small chemical compounds are undergoing clinical trials combined with existing AR antagonists. AR protein elimination by enhanced protein or mRNA degradation is a realistic solution for avoiding AR reactivation during androgen deprivation therapy in prostate cancers.
Collapse
Affiliation(s)
- Jian Huang
- Pathological Diagnosis and Research Center, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Biyun Lin
- Pathological Diagnosis and Research Center, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Benyi Li
- Department of Urology, The University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
77
|
Advances in neuroendocrine prostate cancer research: From model construction to molecular network analyses. J Transl Med 2022; 102:332-340. [PMID: 34937865 DOI: 10.1038/s41374-021-00716-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/25/2021] [Accepted: 11/27/2021] [Indexed: 01/02/2023] Open
Abstract
Prostate cancer is the most common cancer among men and has a high incidence and associated mortality worldwide. It is an androgen-driven disease in which tumor growth is triggered via ligand-mediated signaling through the androgen receptor (AR). Recent evidence suggests that the widespread use of effective AR pathway inhibitors may increase the occurrence of neuroendocrine prostate cancer (NEPC), an aggressive and treatment-resistant AR-negative variant; however, mechanisms controlling NEPC development remain to be elucidated. Various preclinical models have recently been developed to investigate the mechanisms driving the NEPC differentiation. In the present study, we summarized strategies for the development of NEPC models and proposed a novel method for model evaluation, which will help in the timely and accurate identification of NEPC by virtue of its ability to recapitulate the heterogeneity of prostate cancer. Moreover, we discuss the origin and the mechanism of NEPC. The understanding of the regulatory network mediating neuroendocrine differentiation presented in this review could provide valuable insights into the identification of novel drug targets for NEPC as well as into the causes of antiandrogenic drug resistance.
Collapse
|
78
|
METTL14 promotes prostate tumorigenesis by inhibiting THBS1 via an m6A-YTHDF2-dependent mechanism. Cell Death Dis 2022; 8:143. [PMID: 35354789 PMCID: PMC8967870 DOI: 10.1038/s41420-022-00939-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 02/18/2022] [Accepted: 03/08/2022] [Indexed: 11/08/2022]
Abstract
N6-methyladenine (m6A) is the most predominant RNA modification, which has been shown to be related to many types of cancers. However, understanding of its role in prostate cancer (PCa) is largely unknown. Here, we report an upregulation of METTL14 that was correlated with poor prognosis in PCa patients. Functionally, knocking down METTL14 inhibited tumor proliferation both in vitro and in vivo. Mechanically, RNA-seq and MeRIP-seq analyses identified THBS1 as the downstream target of METTL14 in PCa. METTL14 downregulated THBS1 expression in an m6A-dependent manner, which resulted in the recruitment of YTHDF2 to recognize and degrade Thrombospondin 1 (THBS1) mRNA. Thus, our findings revealed that METTL14 acted as an oncogene by inhibiting THBS1 expression via an m6A-YTHDF2-dependent manner. METTL14 could be a potential prognosis marker and a therapeutic target.
Collapse
|
79
|
Ton AT, Foo J, Singh K, Lee J, Kalyta A, Morin H, Perez C, Ban F, Leblanc E, Lallous N, Cherkasov A. Development of VPC-70619, a Small-Molecule N-Myc Inhibitor as a Potential Therapy for Neuroendocrine Prostate Cancer. Int J Mol Sci 2022; 23:ijms23052588. [PMID: 35269731 PMCID: PMC8910697 DOI: 10.3390/ijms23052588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/10/2022] [Accepted: 02/11/2022] [Indexed: 01/18/2023] Open
Abstract
The Myc family of transcription factors are involved in the development and progression of numerous cancers, including prostate cancer (PCa). Under the pressure of androgen receptor (AR)-directed therapies resistance can occur, leading to the lethal form of PCa known as neuroendocrine prostate cancer (NEPC), characterized among other features by N-Myc overexpression. There are no clinically approved treatments for NEPC, translating into poor patient prognosis and survival. Therefore, there is a pressing need to develop novel therapeutic avenues to treat NEPC patients. In this study, we investigate the N-Myc-Max DNA binding domain (DBD) as a potential target for small molecule inhibitors and utilize computer-aided drug design (CADD) approaches to discover prospective hits. Through further exploration and optimization, a compound, VPC-70619, was identified with notable anti-N-Myc potency and strong antiproliferative activity against numerous N-Myc expressing cell lines, including those representing NEPC.
Collapse
|
80
|
Pardo JC, Ruiz de Porras V, Gil J, Font A, Puig-Domingo M, Jordà M. Lipid Metabolism and Epigenetics Crosstalk in Prostate Cancer. Nutrients 2022; 14:851. [PMID: 35215499 PMCID: PMC8874497 DOI: 10.3390/nu14040851] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/27/2022] [Accepted: 02/14/2022] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer (PCa) is the most commonly diagnosed malignant neoplasm in men in the Western world. Localized low-risk PCa has an excellent prognosis thanks to effective local treatments; however, despite the incorporation of new therapeutic strategies, metastatic PCa remains incurable mainly due to disease heterogeneity and the development of resistance to therapy. The mechanisms underlying PCa progression and therapy resistance are multiple and include metabolic reprogramming, especially in relation to lipid metabolism, as well as epigenetic remodelling, both of which enable cancer cells to adapt to dynamic changes in the tumour. Interestingly, metabolism and epigenetics are interconnected. Metabolism can regulate epigenetics through the direct influence of metabolites on epigenetic processes, while epigenetics can control metabolism by directly or indirectly regulating the expression of metabolic genes. Moreover, epidemiological studies suggest an association between a high-fat diet, which can alter the availability of metabolites, and PCa progression. Here, we review the alterations of lipid metabolism and epigenetics in PCa, before focusing on the mechanisms that connect them. We also discuss the influence of diet in this scenario. This information may help to identify prognostic and predictive biomarkers as well as targetable vulnerabilities.
Collapse
Affiliation(s)
- Juan C. Pardo
- Department of Medical Oncology, Catalan Institute of Oncology, University Hospital Germans Trias i Pujol, Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain; (J.C.P.); (A.F.)
- Catalan Institute of Oncology, Badalona Applied Research Group in Oncology (B·ARGO), Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain;
| | - Vicenç Ruiz de Porras
- Catalan Institute of Oncology, Badalona Applied Research Group in Oncology (B·ARGO), Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain;
- Germans Trias i Pujol Research Institute (IGTP), Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain; (J.G.); (M.P.-D.)
| | - Joan Gil
- Germans Trias i Pujol Research Institute (IGTP), Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain; (J.G.); (M.P.-D.)
- Department of Endocrinology and Medicine, CIBERER U747, ISCIII, Research Center for Pituitary Diseases, Hospital Sant Pau, IIB-SPau, Universitat Autònoma de Barcelona, 08041 Barcelona, Spain
| | - Albert Font
- Department of Medical Oncology, Catalan Institute of Oncology, University Hospital Germans Trias i Pujol, Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain; (J.C.P.); (A.F.)
- Catalan Institute of Oncology, Badalona Applied Research Group in Oncology (B·ARGO), Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain;
| | - Manel Puig-Domingo
- Germans Trias i Pujol Research Institute (IGTP), Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain; (J.G.); (M.P.-D.)
- Department of Endocrinology and Nutrition, University Germans Trias i Pujol Hospital, Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain
- Department of Medicine, Autonomous University of Barcelona (UAB), Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain
| | - Mireia Jordà
- Germans Trias i Pujol Research Institute (IGTP), Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain; (J.G.); (M.P.-D.)
| |
Collapse
|
81
|
Wu XC, Yu YZ, Zuo YZ, Song XL, Zhou ZE, Xiao Y, Luo DS, Yan WG, Zhao SC. Identification of UAP1L1 as a critical factor for prostate cancer and underlying molecular mechanism in tumorigenicity. J Transl Med 2022; 20:91. [PMID: 35168617 PMCID: PMC8845250 DOI: 10.1186/s12967-022-03291-0] [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: 08/12/2021] [Accepted: 02/02/2022] [Indexed: 12/24/2022] Open
Abstract
Background Prostate cancer is the second most common cancer in men, and some new target genes are needed to predict the risk of prostate cancer progression and the treatment. Methods In this study, the effects of UAP1L1 (UAP1-like-1) on prostate cancer were investigated by detecting the proliferation, migration, invasion and apoptosis of prostate cancer cells in vitro using MTT, wound healing, Transwell and flow cytometry assay, and the tumor growth in vivo. The downstream genes and pathways of UAP1L1 were explored using Ingenuity Pathway Analysis (IPA), and screened by qRT-PCR and western blot. The effects of CDCA8 on prostate cancer cells were also verified in vitro, which was through detecting the change of proliferation, migration, invasion and apoptosis of prostate cancer cells after CDCA8 knockdown. Results The results indicated that UAP1L1 promoted the proliferation, migration and invasion of prostate cancer cells, which was inhibited by downregulating CDCA8. Furthermore, the promotion of CDCA8 knockdown on cell apoptosis was reduced when UAP1L1 was simultaneously overexpressed. Conclusions In conclusion, the results in this study revealed that UAP1L1 promoted the progression of prostate cancer through the downstream gene CDCA8. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03291-0.
Collapse
Affiliation(s)
- Xing-Cheng Wu
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100720, China
| | - Yu-Zhong Yu
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yu-Zhi Zuo
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100720, China
| | - Xian-Lu Song
- Department of Radiotherapy, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Zhi-En Zhou
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100720, China
| | - Yu Xiao
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Dao-Sheng Luo
- Department of Urology, Dongguan Hospital, Southern Medical University, Guangzhou, China
| | - Wei-Gang Yan
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100720, China.
| | - Shan-Chao Zhao
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
82
|
Liao C, Wang Q, An J, Zhang M, Chen J, Li X, Xiao L, Wang J, Long Q, Liu J, Guan X. SPINKs in Tumors: Potential Therapeutic Targets. Front Oncol 2022; 12:833741. [PMID: 35223512 PMCID: PMC8873584 DOI: 10.3389/fonc.2022.833741] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 01/14/2022] [Indexed: 12/14/2022] Open
Abstract
The serine protease inhibitor Kazal type (SPINK) family includes SPINK1-14 and is the largest branch in the serine protease inhibitor family. SPINKs play an important role in pancreatic physiology and disease, sperm maturation and capacitation, Nager syndrome, inflammation and the skin barrier. Evidence shows that the unregulated expression of SPINK1, 2, 4, 5, 6, 7, and 13 is closely related to human tumors. Different SPINKs exhibit various regulatory modes in different tumors and can be used as tumor prognostic markers. This article reviews the role of SPINK1, 2, 4, 5, 6, 7, and 13 in different human cancer processes and helps to identify new cancer treatment targets.
Collapse
Affiliation(s)
- Chengcheng Liao
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Qian Wang
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, Life Sciences Institute, Zunyi Medical University, Zunyi, China
| | - Jiaxing An
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Minglin Zhang
- Department of Gastroenterology, Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang, China
| | - Jie Chen
- Department of Urology, The Third Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xiaolan Li
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, Life Sciences Institute, Zunyi Medical University, Zunyi, China
| | - Linlin Xiao
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Jiajia Wang
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
| | - Qian Long
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
- *Correspondence: Qian Long, ; Xiaoyan Guan, ; Jianguo Liu,
| | - Jianguo Liu
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
- *Correspondence: Qian Long, ; Xiaoyan Guan, ; Jianguo Liu,
| | - Xiaoyan Guan
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
- *Correspondence: Qian Long, ; Xiaoyan Guan, ; Jianguo Liu,
| |
Collapse
|
83
|
Saigusa N, Hirai H, Tada Y, Kawakita D, Nakaguro M, Tsukahara K, Kano S, Ozawa H, Kondo T, Okami K, Togashi T, Sato Y, Urano M, Kajiwara M, Shimura T, Fushimi C, Shimizu A, Okamoto I, Okada T, Suzuki T, Imanishi Y, Watanabe Y, Sakai A, Ebisumoto K, Sato Y, Honma Y, Yamazaki K, Ueki Y, Hanazawa T, Saito Y, Takahashi H, Ando M, Kohsaka S, Matsuki T, Nagao T. The Role of the EZH2 and H3K27me3 Expression as a Predictor of Clinical Outcomes in Salivary Duct Carcinoma Patients: A Large-Series Study With Emphasis on the Relevance to the Combined Androgen Blockade and HER2-Targeted Therapy. Front Oncol 2022; 11:779882. [PMID: 35186711 PMCID: PMC8850643 DOI: 10.3389/fonc.2021.779882] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/27/2021] [Indexed: 12/15/2022] Open
Abstract
ObjectiveSalivary duct carcinoma (SDC) is a highly aggressive and uncommon tumor arising not only de novo but also in pleomorphic adenoma. Androgen receptor (AR)- and HER2-targeted therapy have recently been introduced for SDC as promising treatment options; however, no predictive biomarkers have yet been established. EZH2 and H3K27me3 are closely linked to the development and progression of various cancers, and EZH2 is also expected to be a desirable therapeutic target. We therefore explored the clinicopathological and prognostic implications of EZH2 and H3K27me3 in a large cohort of SDC patients, focusing on their impact on the therapeutic efficacy of AR- or HER2-targeted therapy.Materials and MethodsThe EZH2 and H3K27me3 immunohistochemical expression and EZH2 Y646 gain-of-function mutation status were examined in 226 SDCs, and the relationship with the clinicopathological factors as well as clinical outcomes were evaluated within the three groups depending on the treatment: AR-targeted (combined androgen blockade with leuprorelin acetate and bicalutamide; 89 cases), HER2-targeted (trastuzumab and docetaxel; 42 cases), and conventional therapy (112 cases).ResultsEZH2 and H3K27me3 were variably immunoreactive in most SDCs. A positive correlation was found between the expression of EZH2 and H3K27me3. The EZH2 expression in the SDC component was significantly higher than that in the pre-existing pleomorphic adenoma component. EZH2 Y646 was not identified in any cases. EZH2-high cases more frequently had an advanced clinical stage and aggressive histological features than EZH2-low cases. An EZH2-high status in patients treated with AR-targeted therapy was associated with a significantly shorter progression-free and overall survival as well as a lower objective response rate and clinical benefit rate. In addition, a H3K27me3-high status in patients treated with AR-targeted therapy was related to a shorter overall survival. Conversely, there was no association between the EZH2 and H3K27me3 expression and the clinical outcomes in the conventional or HER2-targeted therapy groups.ConclusionsA high expression of EZH2 and H3K27me3 in SDC might be a predictor of a poor efficacy of AR-targeted therapy. Our data provide new insights into the role of EZH2 and H3K27me3 in therapeutic strategies for SDC.
Collapse
Affiliation(s)
- Natsuki Saigusa
- Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan
| | - Hideaki Hirai
- Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan
| | - Yuichiro Tada
- Department of Head and Neck Oncology and Surgery, International University of Health and Welfare, Mita Hospital, Tokyo, Japan
| | - Daisuke Kawakita
- Department of Otorhinolaryngology, Head and Neck Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Masato Nakaguro
- Department of Pathology and Laboratory Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Kiyoaki Tsukahara
- Department of Otorhinolaryngology, Head and Neck Surgery, Tokyo Medical University, Tokyo, Japan
| | - Satoshi Kano
- Department of Otolaryngology Head and Neck Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroyuki Ozawa
- Department of Otorhinolaryngology Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Takahito Kondo
- Department of Otorhinolaryngology, Head and Neck Surgery, Tokyo Medical University Hachioji Medical Center, Tokyo, Japan
| | - Kenji Okami
- Department of Otolaryngology Head and Neck Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Takafumi Togashi
- Department of Head and Neck Surgery, Niigata Cancer Center Hospital, Niigata, Japan
| | - Yukiko Sato
- Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Makoto Urano
- Department of Diagnostic Pathology Bantane Hospital Fujita Health University, School of Medicine, Nagoya, Japan
| | - Manami Kajiwara
- Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan
| | - Tomotaka Shimura
- Department of Otolaryngology, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Chihiro Fushimi
- Department of Head and Neck Oncology and Surgery, International University of Health and Welfare, Mita Hospital, Tokyo, Japan
| | - Akira Shimizu
- Department of Otorhinolaryngology, Head and Neck Surgery, Tokyo Medical University, Tokyo, Japan
| | - Isaku Okamoto
- Department of Otorhinolaryngology, Head and Neck Surgery, Tokyo Medical University, Tokyo, Japan
| | - Takuro Okada
- Department of Otorhinolaryngology, Head and Neck Surgery, Tokyo Medical University, Tokyo, Japan
| | - Takayoshi Suzuki
- Department of Otolaryngology Head and Neck Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yorihisa Imanishi
- Department of Otorhinolaryngology Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yoshihiro Watanabe
- Department of Otorhinolaryngology Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Akihiro Sakai
- Department of Otolaryngology Head and Neck Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Koji Ebisumoto
- Department of Otolaryngology Head and Neck Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Yuichiro Sato
- Department of Head and Neck Surgery, Niigata Cancer Center Hospital, Niigata, Japan
| | - Yoshitaka Honma
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Keisuke Yamazaki
- Department of Otolaryngology Head and Neck Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yushi Ueki
- Department of Otolaryngology Head and Neck Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Toyoyuki Hanazawa
- Department of Otolaryngology, Head and Neck Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yuki Saito
- Department of Otolaryngology - Head and Neck Surgery, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hideaki Takahashi
- Department of Otorhinolaryngology, Head and Neck Surgery, Yokohama City University, School of Medicine, Yokohama, Japan
| | - Mizuo Ando
- Department of Otolaryngology-Head & Neck Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shinji Kohsaka
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Takashi Matsuki
- Department of Otorhinolaryngology, Head and Neck Surgery, Kitasato University School of Medicine, Kanagawa, Japan
| | - Toshitaka Nagao
- Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan
- *Correspondence: Toshitaka Nagao,
| |
Collapse
|
84
|
Merkens L, Sailer V, Lessel D, Janzen E, Greimeier S, Kirfel J, Perner S, Pantel K, Werner S, von Amsberg G. Aggressive variants of prostate cancer: underlying mechanisms of neuroendocrine transdifferentiation. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:46. [PMID: 35109899 PMCID: PMC8808994 DOI: 10.1186/s13046-022-02255-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/13/2022] [Indexed: 12/14/2022]
Abstract
Prostate cancer is a hormone-driven disease and its tumor cell growth highly relies on increased androgen receptor (AR) signaling. Therefore, targeted therapy directed against androgen synthesis or AR activation is broadly used and continually improved. However, a subset of patients eventually progresses to castration-resistant disease. To date, various mechanisms of resistance have been identified including the development of AR-independent aggressive variant prostate cancer based on neuroendocrine transdifferentiation (NED). Here, we review the highly complex processes contributing to NED. Genetic, epigenetic, transcriptional aberrations and posttranscriptional modifications are highlighted and the potential interplay of the different factors is discussed. Background Aggressive variant prostate cancer (AVPC) with traits of neuroendocrine differentiation emerges in a rising number of patients in recent years. Among others, advanced therapies targeting the androgen receptor axis have been considered causative for this development. Cell growth of AVPC often occurs completely independent of the androgen receptor signal transduction pathway and cells have mostly lost the typical cellular features of prostate adenocarcinoma. This complicates both diagnosis and treatment of this very aggressive disease. We believe that a deeper understanding of the complex molecular pathological mechanisms contributing to transdifferentiation will help to improve diagnostic procedures and develop effective treatment strategies. Indeed, in recent years, many scientists have made important contributions to unravel possible causes and mechanisms in the context of neuroendocrine transdifferentiation. However, the complexity of the diverse molecular pathways has not been captured completely, yet. This narrative review comprehensively highlights the individual steps of neuroendocrine transdifferentiation and makes an important contribution in bringing together the results found so far.
Collapse
Affiliation(s)
- Lina Merkens
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
| | - Verena Sailer
- Institute of Pathology, University of Luebeck and University Hospital Schleswig-Holstein, Campus Luebeck, Ratzeburger Allee 160, 23538, Luebeck, Germany
| | - Davor Lessel
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Ella Janzen
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Sarah Greimeier
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Jutta Kirfel
- Institute of Pathology, University of Luebeck and University Hospital Schleswig-Holstein, Campus Luebeck, Ratzeburger Allee 160, 23538, Luebeck, Germany
| | - Sven Perner
- Institute of Pathology, University of Luebeck and University Hospital Schleswig-Holstein, Campus Luebeck, Ratzeburger Allee 160, 23538, Luebeck, Germany.,Pathology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Klaus Pantel
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.,European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Stefan Werner
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.,Mildred Scheel Cancer Career Center Hamburg HaTRiCs4, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gunhild von Amsberg
- Department of Hematology and Oncology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.,Martini-Klinik, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| |
Collapse
|
85
|
Owens JL, Beketova E, Liu S, Shen Q, Pawar JS, Asberry AM, Yang J, Deng X, Elzey BD, Ratliff TL, Cheng L, Choo CR, Citrin DE, Polascik TJ, Wang B, Huang J, Li C, Wan J, Hu CD. Targeting protein arginine methyltransferase 5 (PRMT5) suppresses radiation-induced neuroendocrine differentiation and sensitizes prostate cancer cells to radiation. Mol Cancer Ther 2022; 21:448-459. [PMID: 35027481 DOI: 10.1158/1535-7163.mct-21-0103] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 10/17/2021] [Accepted: 01/04/2022] [Indexed: 11/16/2022]
Abstract
Prostate cancer remains the second leading cause of cancer death among American men. Radiation therapy (RT) is a potentially curative treatment for localized prostate cancer, and failure to control localized disease contributes to the majority of prostate cancer deaths. Neuroendocrine differentiation (NED) in prostate cancer, a process by which prostate adenocarcinoma cells transdifferentiate into neuroendocrine-like (NE-like) cells, is an emerging mechanism of resistance to cancer therapies and contributes to disease progression. NED also occurs in response to treatment to promote the development of treatment-induced neuroendocrine prostate cancer (NEPC), a highly-aggressive and terminal stage disease. We previously demonstrated that by mimicking clinical RT protocol, fractionated ionizing radiation (FIR) induces prostate cancer cells to undergo NED in vitro and in vivo. Here, we performed transcriptomic analysis and confirmed that FIR-induced NE-like cells share some features of clinical NEPC, suggesting that FIR-induced NED represents a clinically-relevant model. Further, we demonstrated that protein arginine methyltransferase 5 (PRMT5), a master epigenetic regulator of the DNA damage response and a putative oncogene in prostate cancer, along with its cofactors pICln and MEP50, mediate FIR-induced NED. Knockdown of PRMT5, pICln, or MEP50 during FIR-inhibited NED sensitized prostate cancer cells to radiation. Significantly, PRMT5 knockdown in prostate cancer xenograft tumors in mice during FIR prevented NED, enhanced tumor killing, significantly reduced and delayed tumor recurrence, and prolonged overall survival. Collectively, our results demonstrate that PRMT5 promotes FIR-induced NED and suggests that targeting PRMT5 may be a novel and effective radiosensitization approach for prostate cancer RT.
Collapse
Affiliation(s)
- Jake L Owens
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University West Lafayette
| | - Elena Beketova
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University West Lafayette
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine
| | - Qi Shen
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University West Lafayette
| | - Jogendra Singh Pawar
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University West Lafayette
| | - Andrew M Asberry
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University West Lafayette
| | - Jie Yang
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University West Lafayette
| | - Xuehong Deng
- Medicinal Chemistry and Molecular Pharmacolog, Purdue University West Lafayette
| | - Bennett D Elzey
- Department of Comparative Pathobiology, Purdue University West Lafayette
| | - Timothy L Ratliff
- Comparative Pathobiology and the Center for Cancer Research, Purdue University West Lafayette
| | - Liang Cheng
- Pathology and Laboratory Medicine, Indiana University School of Medicine
| | | | | | | | - Bangchen Wang
- Department of Pathology, Duke University School of Medicine
| | - Jiaoti Huang
- Department of Pathology, Duke University School of Medicine
| | | | - Jun Wan
- Medical and Molecular Genetics, Indiana University School of Medicine
| | - Chang-Deng Hu
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University West Lafayette
| |
Collapse
|
86
|
Song H, Weinstein HNW, Allegakoen P, Wadsworth MH, Xie J, Yang H, Castro EA, Lu KL, Stohr BA, Feng FY, Carroll PR, Wang B, Cooperberg MR, Shalek AK, Huang FW. Single-cell analysis of human primary prostate cancer reveals the heterogeneity of tumor-associated epithelial cell states. Nat Commun 2022; 13:141. [PMID: 35013146 PMCID: PMC8748675 DOI: 10.1038/s41467-021-27322-4] [Citation(s) in RCA: 124] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 10/29/2021] [Indexed: 12/31/2022] Open
Abstract
Prostate cancer is the second most common malignancy in men worldwide and consists of a mixture of tumor and non-tumor cell types. To characterize the prostate cancer tumor microenvironment, we perform single-cell RNA-sequencing on prostate biopsies, prostatectomy specimens, and patient-derived organoids from localized prostate cancer patients. We uncover heterogeneous cellular states in prostate epithelial cells marked by high androgen signaling states that are enriched in prostate cancer and identify a population of tumor-associated club cells that may be associated with prostate carcinogenesis. ERG-negative tumor cells, compared to ERG-positive cells, demonstrate shared heterogeneity with surrounding luminal epithelial cells and appear to give rise to common tumor microenvironment responses. Finally, we show that prostate epithelial organoids harbor tumor-associated epithelial cell states and are enriched with distinct cell types and states from their parent tissues. Our results provide diagnostically relevant insights and advance our understanding of the cellular states associated with prostate carcinogenesis.
Collapse
Affiliation(s)
- Hanbing Song
- grid.266102.10000 0001 2297 6811Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Hannah N. W. Weinstein
- grid.266102.10000 0001 2297 6811Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Paul Allegakoen
- grid.266102.10000 0001 2297 6811Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Marc H. Wadsworth
- grid.116068.80000 0001 2341 2786The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139 USA ,grid.116068.80000 0001 2341 2786Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA 02139 USA ,grid.116068.80000 0001 2341 2786Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139 USA ,grid.116068.80000 0001 2341 2786Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139 USA ,grid.66859.340000 0004 0546 1623Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142 USA
| | - Jamie Xie
- grid.266102.10000 0001 2297 6811Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Heiko Yang
- grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Department of Urology, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Ethan A. Castro
- grid.266102.10000 0001 2297 6811Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Kevin L. Lu
- grid.266102.10000 0001 2297 6811Department of Pathology, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Bradley A. Stohr
- grid.266102.10000 0001 2297 6811Department of Pathology, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Felix Y. Feng
- grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Department of Urology, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Departments of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Peter R. Carroll
- grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Department of Urology, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Bruce Wang
- grid.266102.10000 0001 2297 6811Division of Gastroenterology, Department of Medicine, University of California, San Francisco, CA 94143 USA
| | - Matthew R. Cooperberg
- grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Department of Urology, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.410372.30000 0004 0419 2775Division of Hematology and Oncology, Department of Medicine, San Francisco Veterans Affairs Medical Center, San Francisco, CA 94121 USA
| | - Alex K. Shalek
- grid.116068.80000 0001 2341 2786The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139 USA ,grid.116068.80000 0001 2341 2786Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA 02139 USA ,grid.116068.80000 0001 2341 2786Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139 USA ,grid.116068.80000 0001 2341 2786Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139 USA ,grid.66859.340000 0004 0546 1623Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142 USA
| | - Franklin W. Huang
- grid.266102.10000 0001 2297 6811Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.410372.30000 0004 0419 2775Division of Hematology and Oncology, Department of Medicine, San Francisco Veterans Affairs Medical Center, San Francisco, CA 94121 USA
| |
Collapse
|
87
|
Gravemeyer J, Spassova I, Verhaegen ME, Dlugosz AA, Hoffmann D, Lange A, Becker JC. DNA-methylation patterns imply a common cellular origin of virus- and UV-associated Merkel cell carcinoma. Oncogene 2022; 41:37-45. [PMID: 34667274 PMCID: PMC8724008 DOI: 10.1038/s41388-021-02064-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/22/2021] [Accepted: 10/04/2021] [Indexed: 02/07/2023]
Abstract
Merkel cell carcinoma (MCC) is a neuroendocrine tumor either induced by integration of the Merkel cell polyomavirus into the cell genome or by accumulation of UV-light-associated mutations (VP-MCC and UV-MCC). Whether VP- and UV-MCC have the same or different cellular origins is unclear; with mesenchymal or epidermal origins discussed. DNA-methylation patterns have a proven utility in determining cellular origins of cancers. Therefore, we used this approach to uncover evidence regarding the cell of origin of classical VP- and UV-MCC cell lines, i.e., cell lines with a neuroendocrine growth pattern (n = 9 and n = 4, respectively). Surprisingly, we observed high global similarities in the DNA-methylation of UV- and VP-MCC cell lines. CpGs of lower methylation in VP-MCC cell lines were associated with neuroendocrine marker genes such as SOX2 and INSM1, or linked to binding sites of EZH2 and SUZ12 of the polycomb repressive complex 2, i.e., genes with an impact on carcinogenesis and differentiation of neuroendocrine cancers. Thus, the observed differences appear to be rooted in viral compared to mutation-driven carcinogenesis rather than distinct cells of origin. To test this hypothesis, we used principal component analysis, to compare DNA-methylation data from different epithelial and non-epithelial neuroendocrine cancers and established a scoring model for epithelial and neuroendocrine characteristics. Subsequently, we applied this scoring model to the DNA-methylation data of the VP- and UV-MCC cell lines, revealing that both clearly scored as epithelial cancers. In summary, our comprehensive analysis of DNA-methylation suggests a common epithelial origin of UV- and VP-MCC cell lines.
Collapse
Affiliation(s)
- Jan Gravemeyer
- Translational Skin Cancer Research (TSCR), University Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK) & German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ivelina Spassova
- Translational Skin Cancer Research (TSCR), University Duisburg-Essen, Essen, Germany
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | | | - Andrzej A Dlugosz
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Daniel Hoffmann
- Bioinformatics & Computational Biophysics, University Duisburg-Essen, Essen, Germany
| | - Anja Lange
- Bioinformatics & Computational Biophysics, University Duisburg-Essen, Essen, Germany
| | - Jürgen C Becker
- Translational Skin Cancer Research (TSCR), University Duisburg-Essen, Essen, Germany.
- German Cancer Consortium (DKTK) & German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Department of Dermatology, University Hospital Essen, Essen, Germany.
| |
Collapse
|
88
|
Deng L, Li C, He Q, Huang C, Chen Q, Zhang S, Wang L, Gan Y, Long Z. Superselective Prostate Artery Embolization for Treatment of Severe Haematuria Secondary to Rapid Progression of Treatment-Induced Neuroendocrine Prostate Cancer: A Case Report. Onco Targets Ther 2022; 15:67-75. [PMID: 35082500 PMCID: PMC8786387 DOI: 10.2147/ott.s345193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/11/2022] [Indexed: 11/23/2022] Open
Abstract
Background Treatment-induced neuroendocrine prostate cancer (t-NEPC) represents a highly aggressive subtype of castration-resistant prostate cancer that commonly arises from prostate adenocarcinoma (AdPC) after continuous androgen deprivation therapy (ADT). However, current treatments for t-NEPC are limited and far from satisfactory. According to our limited knowledge, report regarding the management of t-NEPC related hemorrhage is rare. Here, we report a case of t-NEPC formation after chronic hormonal therapy accompanying with severe bleeding in primary tumor and share our experiences to deal with the severe hematuria resulting from the progression of t-NEPC tumor. Case Presentation An 80-year-old man with a significantly high prostate-specific antigen was diagnosed via pathology as advanced AdPC due to multiple bone metastases. He then received ADT including bicalutamide and goserelin. After 20 months of stable disease, the cancer rapidly progressed and presented with severe gross hematuria caused by bleeding of the primary tumor. The histopathologic analysis of a secondary biopsy of the primary tumor confirmed neuroendocrine prostate cancer, and subsequent genetic testing revealed germ-line mutations in the RB1 and FOXA1. To control the bleeding and relieve symptoms, the patient was treated with superselective prostate artery embolization (PAE). After the left internal pudendal artery and the right prostatic artery were embolized, hematuria was quickly alleviated and disappeared. However, the patient was not a suitable candidate to platinum-based chemotherapy due to weak constitution. Goserelin was continuously applied to maintain castration level of serum testosterone. Meanwhile, palliative radiotherapy to the prostate tumor, high-risk lymph node drainage areas (including iliac and para-aortic lymph nodes, internal iliac lymph nodes, presacral lymph nodes and obturator nerve lymph nodes) and bone metastases (right sacroiliac joint and thoracic vertebra) was performed and relieved the pain. Unfortunately, this patient eventually died of cachexia and multiple organ failure nearly 27 months after initial diagnosis. Conclusion To treat severe hematuria caused by progression of t-NEPC, superselective PAE may be a rapid and efficient way to stop bleeding.
Collapse
Affiliation(s)
- Liang Deng
- Andrology Center, Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, 410013, People’s Republic of China
| | - Chao Li
- Andrology Center, Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, 410013, People’s Republic of China
| | - Qiangrong He
- Andrology Center, Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, 410013, People’s Republic of China
| | - Chenghui Huang
- Department of Medical Oncology, The Third Xiangya Hospital of Central South University, Changsha, 410013, People’s Republic of China
| | - Qian Chen
- Department of Pathology, The Third Xiangya Hospital of Central South University, Changsha, 410013, People’s Republic of China
| | - Shengwang Zhang
- Department of Radiation Oncology, The Third Xiangya Hospital of Central South University, Changsha, 410013, People’s Republic of China
| | - Long Wang
- Andrology Center, Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, 410013, People’s Republic of China
| | - Yu Gan
- Department of Urology, Xiangya Hospital of Central South University, Changsha, 410008, People’s Republic of China
- Correspondence: Yu Gan, Department of Urology, Xiangya Hospital of Central South University, 87 Xiang Road, Changsha, Hunan, People’s Republic of China, Tel +86 15111140206, Fax +86 73184327332, Email
| | - Zhi Long
- Andrology Center, Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, 410013, People’s Republic of China
- Zhi Long, Andrology Center, Department of Urology, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, Hunan, People’s Republic of China, Tel +86 13755076226, Fax +86 73188618028 Email
| |
Collapse
|
89
|
Cheng Q, Butler W, Zhou Y, Zhang H, Tang L, Perkinson K, Chen X, Jiang X“S, McCall SJ, Inman BA, Huang J. Pre-existing Castration-resistant Prostate Cancer–like Cells in Primary Prostate Cancer Promote Resistance to Hormonal Therapy. Eur Urol 2022; 81:446-455. [PMID: 35058087 PMCID: PMC9018600 DOI: 10.1016/j.eururo.2021.12.039] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 12/01/2021] [Accepted: 12/31/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Hormonal therapy targeting the androgen receptor inhibits prostate cancer (PCa), but the tumor eventually recurs as castration-resistant prostate cancer (CRPC). OBJECTIVE To understand the mechanisms by which subclones within early PCa develop into CRPC. DESIGN, SETTING, AND PARTICIPANTS We isolated epithelial cells from fresh human PCa cases, including primary adenocarcinoma, locally recurrent CRPC, and metastatic CRPC, and utilized single-cell RNA sequencing to identify subpopulations destined to become either CRPC-adeno or small cell neuroendocrine carcinoma (SCNC). OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS We revealed dynamic transcriptional reprogramming that promotes disease progression among 23226 epithelial cells using single-cell RNA sequencing, and validated subset-specific progression using immunohistochemistry and large cohorts of publically available genomic data. RESULTS AND LIMITATIONS We identified a small fraction of highly plastic CRPC-like cells in hormone-naïve early PCa and demonstrated its correlation with biochemical recurrence and distant metastasis, independent of clinical characteristics. We show that progression toward castration resistance was initiated from subtype-specific lineage plasticity and clonal expansion of pre-existing neuroendocrine and CRPC-like cells in early PCa. CONCLUSIONS CRPC-like cells are present early in the development of PCa and are not exclusively the result of acquired evolutionary selection during androgen deprivation therapy. The lethal CRPC and SCNC phenotypes should be targeted earlier in the disease course of patients with PCa. PATIENT SUMMARY Here, we report the presence of pre-existing castration-resistant prostate cancer (CRPC)-like cells in primary prostate cancer, which represents a novel castration-resistant mechanism different from the adaptation mechanism after androgen deprivation therapy (ADT). Patients whose tumors harbor increased pre-existing neuroendocrine and CRPC-like cells may become rapidly resistant to ADT and may require aggressive early intervention.
Collapse
|
90
|
Hu CY, Wu KY, Lin TY, Chen CC. The Crosstalk of Long Non-Coding RNA and MicroRNA in Castration-Resistant and Neuroendocrine Prostate Cancer: Their Interaction and Clinical Importance. Int J Mol Sci 2021; 23:ijms23010392. [PMID: 35008817 PMCID: PMC8745162 DOI: 10.3390/ijms23010392] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/02/2021] [Accepted: 12/28/2021] [Indexed: 12/18/2022] Open
Abstract
Prostate cancer is featured by its heterogeneous nature, which indicates a different prognosis. Castration-resistant prostate cancer (CRPC) is a hallmark of the treatment-refractory stage, and the median survival of patients is only within two years. Neuroendocrine prostate cancer (NEPC) is an aggressive variant that arises from de novo presentation of small cell carcinoma or treatment-related transformation with a median survival of 1–2 years from the time of diagnosis. The epigenetic regulators, such as long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), have been proven involved in multiple pathologic mechanisms of CRPC and NEPC. LncRNAs can act as competing endogenous RNAs to sponge miRNAs that would inhibit the expression of their targets. After that, miRNAs interact with the 3’ untranslated region (UTR) of target mRNAs to repress the step of translation. These interactions may modulate gene expression and influence cancer development and progression. Otherwise, epigenetic regulators and genetic mutation also promote neuroendocrine differentiation and cancer stem-like cell formation. This step may induce neuroendocrine prostate cancer development. This review aims to provide an integrated, synthesized overview under current evidence to elucidate the crosstalk of lncRNAs with miRNAs and their influence on castration resistance or neuroendocrine differentiation of prostate cancer. Notably, we also discuss the mechanisms of lncRNA–miRNA interaction in androgen receptor-independent prostate cancer, such as growth factors, oncogenic signaling pathways, cell cycle dysregulation, and cytokines or other transmembrane proteins. Conclusively, we underscore the potential of these communications as potential therapeutic targets in the future.
Collapse
Affiliation(s)
- Che-Yuan Hu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan;
- Department of Urology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan;
| | - Kuan-Yu Wu
- Department of Urology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan;
| | - Tsung-Yen Lin
- Department of Urology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan;
- Division of Urology, Department of Surgery, Dou-Liou Branch, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Yunlin 640, Taiwan
- Correspondence: (T.-Y.L.); (C.-C.C.); Tel.: +886-6235-3535 (ext. 5251) (T.-Y.L.); +886-5276-5041 (ext. 7521) (C.-C.C.)
| | - Chien-Chin Chen
- Department of Pathology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 600, Taiwan
- Department of Cosmetic Science, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan
- Correspondence: (T.-Y.L.); (C.-C.C.); Tel.: +886-6235-3535 (ext. 5251) (T.-Y.L.); +886-5276-5041 (ext. 7521) (C.-C.C.)
| |
Collapse
|
91
|
Singh N, Ramnarine VR, Song JH, Pandey R, Padi SKR, Nouri M, Olive V, Kobelev M, Okumura K, McCarthy D, Hanna MM, Mukherjee P, Sun B, Lee BR, Parker JB, Chakravarti D, Warfel NA, Zhou M, Bearss JJ, Gibb EA, Alshalalfa M, Karnes RJ, Small EJ, Aggarwal R, Feng F, Wang Y, Buttyan R, Zoubeidi A, Rubin M, Gleave M, Slack FJ, Davicioni E, Beltran H, Collins C, Kraft AS. The long noncoding RNA H19 regulates tumor plasticity in neuroendocrine prostate cancer. Nat Commun 2021; 12:7349. [PMID: 34934057 PMCID: PMC8692330 DOI: 10.1038/s41467-021-26901-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 10/22/2021] [Indexed: 12/15/2022] Open
Abstract
Neuroendocrine (NE) prostate cancer (NEPC) is a lethal subtype of castration-resistant prostate cancer (PCa) arising either de novo or from transdifferentiated prostate adenocarcinoma following androgen deprivation therapy (ADT). Extensive computational analysis has identified a high degree of association between the long noncoding RNA (lncRNA) H19 and NEPC, with the longest isoform highly expressed in NEPC. H19 regulates PCa lineage plasticity by driving a bidirectional cell identity of NE phenotype (H19 overexpression) or luminal phenotype (H19 knockdown). It contributes to treatment resistance, with the knockdown of H19 re-sensitizing PCa to ADT. It is also essential for the proliferation and invasion of NEPC. H19 levels are negatively regulated by androgen signaling via androgen receptor (AR). When androgen is absent SOX2 levels increase, driving H19 transcription and facilitating transdifferentiation. H19 facilitates the PRC2 complex in regulating methylation changes at H3K27me3/H3K4me3 histone sites of AR-driven and NEPC-related genes. Additionally, this lncRNA induces alterations in genome-wide DNA methylation on CpG sites, further regulating genes associated with the NEPC phenotype. Our clinical data identify H19 as a candidate diagnostic marker and predictive marker of NEPC with elevated H19 levels associated with an increased probability of biochemical recurrence and metastatic disease in patients receiving ADT. Here we report H19 as an early upstream regulator of cell fate, plasticity, and treatment resistance in NEPC that can reverse/transform cells to a treatable form of PCa once therapeutically deactivated.
Collapse
MESH Headings
- Androgen Antagonists/therapeutic use
- Animals
- Benzamides/pharmacology
- Benzamides/therapeutic use
- Biomarkers, Tumor/metabolism
- Carcinoma, Neuroendocrine/diagnosis
- Carcinoma, Neuroendocrine/drug therapy
- Carcinoma, Neuroendocrine/genetics
- Carcinoma, Neuroendocrine/pathology
- Cell Line, Tumor
- Cell Lineage/genetics
- Cell Nucleus/metabolism
- Cell Plasticity/genetics
- Cell Proliferation/genetics
- Cohort Studies
- DNA Methylation/genetics
- Disease Models, Animal
- Drug Resistance, Neoplasm/genetics
- Epigenesis, Genetic/drug effects
- Gene Expression Regulation, Neoplastic
- Genome, Human
- Histones/metabolism
- Humans
- Male
- Neoplasm Grading
- Neoplasm Invasiveness
- Neoplastic Stem Cells/metabolism
- Nitriles/pharmacology
- Nitriles/therapeutic use
- Organoids/metabolism
- Organoids/pathology
- Phenylthiohydantoin/pharmacology
- Phenylthiohydantoin/therapeutic use
- Phylogeny
- Polycomb Repressive Complex 2/metabolism
- Promoter Regions, Genetic/genetics
- Prostatic Neoplasms/diagnosis
- Prostatic Neoplasms/drug therapy
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/pathology
- Protein Isoforms/genetics
- Protein Isoforms/metabolism
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Receptors, Androgen/metabolism
- SOXB1 Transcription Factors/metabolism
- Transcription, Genetic/drug effects
Collapse
Affiliation(s)
- Neha Singh
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, 85724, USA
| | - Varune R Ramnarine
- Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
- Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, MA, 02215, USA
| | - Jin H Song
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, 85724, USA
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, 85724, USA
| | - Ritu Pandey
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, 85724, USA
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, 85724, USA
| | - Sathish K R Padi
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, 85724, USA
- Department of Molecular Biology and Biophysics, UConn Health Center, Farmington, CT, 06030, USA
| | - Mannan Nouri
- Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Virginie Olive
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, 85724, USA
- Department of Medicine, University of Arizona, Tucson, AZ, 85724, USA
| | - Maxim Kobelev
- Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Koichi Okumura
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, 85724, USA
- Department of Physiology, University of Arizona, Tucson, AZ, 85724, USA
| | - David McCarthy
- Ribomed Biotechnologies, Inc., 8821N. 7th St. STE 300, Phoenix, AZ, 85020, USA
| | - Michelle M Hanna
- Ribomed Biotechnologies, Inc., 8821N. 7th St. STE 300, Phoenix, AZ, 85020, USA
| | - Piali Mukherjee
- Epigenomics Core Facility, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Belinda Sun
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, 85724, USA
| | - Benjamin R Lee
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, 85724, USA
| | - J Brandon Parker
- Department of Obstetrics and Gynecology, Division of Reproductive Science in Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Debabrata Chakravarti
- Department of Obstetrics and Gynecology, Division of Reproductive Science in Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Noel A Warfel
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, 85724, USA
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, 85724, USA
| | - Muhan Zhou
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, 85724, USA
| | - Jeremiah J Bearss
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, 85724, USA
| | - Ewan A Gibb
- Decipher Biosciences, Inc, Vancouver, BC, Canada
| | - Mohammed Alshalalfa
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94158, USA
| | - R Jefferey Karnes
- Department of Urology, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA
| | - Eric J Small
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Rahul Aggarwal
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Felix Feng
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Yuzhuo Wang
- Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Ralph Buttyan
- Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Amina Zoubeidi
- Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Mark Rubin
- Department for BioMedical Research, University of Bern, 3008, Bern, Switzerland
| | - Martin Gleave
- Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Frank J Slack
- Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, MA, 02215, USA
| | | | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, 44 Binney St, Boston, MA, 02115, USA
| | - Colin Collins
- Vancouver Prostate Centre & Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada.
| | - Andrew S Kraft
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, 85724, USA.
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, 85724, USA.
| |
Collapse
|
92
|
Slabáková E, Kahounová Z, Procházková J, Souček K. Regulation of Neuroendocrine-like Differentiation in Prostate Cancer by Non-Coding RNAs. Noncoding RNA 2021; 7:ncrna7040075. [PMID: 34940756 PMCID: PMC8704250 DOI: 10.3390/ncrna7040075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/18/2021] [Accepted: 11/29/2021] [Indexed: 12/21/2022] Open
Abstract
Neuroendocrine prostate cancer (NEPC) represents a variant of prostate cancer that occurs in response to treatment resistance or, to a much lesser extent, de novo. Unravelling the molecular mechanisms behind transdifferentiation of cancer cells to neuroendocrine-like cancer cells is essential for development of new treatment opportunities. This review focuses on summarizing the role of small molecules, predominantly microRNAs, in this phenomenon. A published literature search was performed to identify microRNAs, which are reported and experimentally validated to modulate neuroendocrine markers and/or regulators and to affect the complex neuroendocrine phenotype. Next, available patients’ expression datasets were surveyed to identify deregulated microRNAs, and their effect on NEPC and prostate cancer progression is summarized. Finally, possibilities of miRNA detection and quantification in body fluids of prostate cancer patients and their possible use as liquid biopsy in prostate cancer monitoring are discussed. All the addressed clinical and experimental contexts point to an association of NEPC with upregulation of miR-375 and downregulation of miR-34a and miR-19b-3p. Together, this review provides an overview of different roles of non-coding RNAs in the emergence of neuroendocrine prostate cancer.
Collapse
|
93
|
Kanayama M, Luo J. Delineating the Molecular Events Underlying Development of Prostate Cancer Variants with Neuroendocrine/Small Cell Carcinoma Characteristics. Int J Mol Sci 2021; 22:12742. [PMID: 34884545 PMCID: PMC8657721 DOI: 10.3390/ijms222312742] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/17/2021] [Accepted: 11/19/2021] [Indexed: 12/18/2022] Open
Abstract
The treatment landscape of prostate cancer has changed dramatically following the advent of novel systemic therapies, most of which target the androgen receptor (AR). Agents such as abiraterone, enzalutamide, apalutamide, darolutamide were designed to further suppress androgen receptor signaling following gonadal suppression achieved by first-line androgen deprivation therapies. These potent AR targeting agents are increasingly used in the earlier stages of the disease spectrum with the goal of delaying disease progression and extending survival. Although these therapies are effective in controlling prostate tumors dependent on or addicted to AR signaling, prostate tumors surviving the onslaught of potent treatments may evolve and develop drug resistance. A substantial proportion of treatment failures can be explained by the development of treatment-induced aggressive prostate cancer variants such as neuroendocrine/small cell carcinoma. These emerging disease entities demand detailed characterization and precise definitions. We postulate that these treatment-induced prostate cancer entities should be defined molecularly to overcome the drawbacks associated with the current clinical and pathological definitions. A precise molecular definition conforms with current knowledge on the molecular evolution of this disease entity and will enable early detection and early intervention.
Collapse
Affiliation(s)
- Mayuko Kanayama
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
| | | |
Collapse
|
94
|
Wen YC, Liu YN, Yeh HL, Chen WH, Jiang KC, Lin SR, Huang J, Hsiao M, Chen WY. TCF7L1 regulates cytokine response and neuroendocrine differentiation of prostate cancer. Oncogenesis 2021; 10:81. [PMID: 34799554 PMCID: PMC8604986 DOI: 10.1038/s41389-021-00371-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 09/01/2021] [Accepted: 11/03/2021] [Indexed: 01/07/2023] Open
Abstract
Neuroendocrine differentiation (NED) is associated with WNT signaling activation and can be significantly observed after failure of androgen-deprivation therapy (ADT) for prostatic adenocarcinomas. Cytokine signaling is stimulated in NED prostate cancer; however, how ADT-upregulated WNT signaling promotes activation of cytokine signaling and contributes to NED of prostate cancer is poorly understood. In this study, we identified ADT-mediated upregulation of transcription factor 7 like 1 (TCF7L1), which increases the cytokine response and enhances NED of prostate cancer through interleukin (IL)-8/C-X-C motif chemokine receptor type 2 (CXCR2) signaling activation. ADT induced the secretion of WNT4 which upon engagement of TCF7L1 in prostate cancer cells, enhanced IL-8 and CXCR2 expressions. TCF7L1 directly binds to the regulatory sequence region of IL-8 and CXCR2 through WNT4 activation, thus upregulating IL-8/CXCR2 signaling-driven NED and cell motility. Analysis of prostate tissue samples collected from small-cell neuroendocrine prostate cancer (SCPC) and castration-resistant prostate cancer (CRPC) tumors showed an increased intensity of nuclear TCF7L1 associated with CXCR2. Our results suggest that induction of WNT4/TCF7L1 results in increased NED and malignancy in prostate cancer that is linked to dysregulation of androgen receptor signaling and activation of the IL-8/CXCR2 pathway.
Collapse
Affiliation(s)
- Yu-Ching Wen
- Department of Urology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan
| | - Yen-Nien Liu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Hsiu-Lien Yeh
- General Education Development Center, Hsin Sheng Junior College of Medical Care and Management, Taoyuan, Taiwan
| | - Wei-Hao Chen
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Kuo-Ching Jiang
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Shian-Ren Lin
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Jiaoti Huang
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Wei-Yu Chen
- Department of Pathology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan. .,Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
95
|
CKB inhibits epithelial-mesenchymal transition and prostate cancer progression by sequestering and inhibiting AKT activation. Neoplasia 2021; 23:1147-1165. [PMID: 34706306 PMCID: PMC8551525 DOI: 10.1016/j.neo.2021.09.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 09/17/2021] [Indexed: 12/26/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) contributes to tumor invasion, metastasis and drug resistance. AKT activation is key in a number of cellular processes. While many positive regulators for either EMT or AKT activation have been reported, few negative regulators are established. Through kinase cDNA screen, we identified brain-type creatine kinase (CKB or BCK) as a potent suppressor for both. As a ubiquitously expressed kinase in normal tissues, CKB is significantly downregulated in several solid cancer types. Lower CKB expression is significantly associated with worse prognosis. Phenotypically, CKB overexpression suppresses, while its silencing promotes, EMT and cell migration, xenograft tumor growth and metastasis of prostate cancer cells. AKT activation is one of the most prominent signaling events upon CKB silencing in prostate cancer cells, which is in line with prostate cancer TCGA data. EMT enhanced by CKB silencing is abolished by AKT inhibition. Mechanistically, CKB interacts with AKT and sequestrates it from activation by mTOR. We further elucidated that an 84aa fragment at C-terminus of CKB protein interacts with AKT's PH domain. Ectopic expression of the 84aa CKB fragment inhibits AKT activation, EMT and cell proliferation. Interestingly, molecular dynamics simulation on crystal structures of AKT and CKB independently demonstrates that AKT's PH domain and CKB's 84aa fragment establish their major interaction interface. In summary, we have discovered CKB as a negative regulator of EMT and AKT activation, revealing a new mode of their regulation . We have also demonstrated that CKB downregulation is a poor prognosticator, which is sufficient to promote prostate cancer progression.
Collapse
|
96
|
Oey O, Ghaffari M, Li JJ, Hosseini-Beheshti E. Application of extracellular vesicles in the diagnosis and treatment of prostate cancer: Implications for clinical practice. Crit Rev Oncol Hematol 2021; 167:103495. [PMID: 34655743 DOI: 10.1016/j.critrevonc.2021.103495] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 07/12/2021] [Accepted: 09/08/2021] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EV) are cell-derived lipid bilayer-delimited structures providing an important means of intercellular communication. Recent studies have shown that EV, particularly exosomes and large-oncosomes contain miRNA and proteins crucial in prostate cancer (PCa) progression, metastasis and treatment resistance. This includes not just EV released from PCa cells, but also from other cells in the tumor microenvironment. PCa patient derived EV have a unique composition compared to healthy and benign prostatic diseases. As such, EV show promise as diagnostic liquid biopsy biomarkers, both as an adjunct and alternative to the invasive current gold-standard. EV could also be utilized to stratify patients' risk and predict response to hormonal, chemo, immune- and targeted therapy, which will direct future treatment decisions in PCa. We present a summary of the current evidence on the role of EV in PCa and the application of EV in PCa diagnosis and treatment to optimize patient outcomes.
Collapse
Affiliation(s)
- Oliver Oey
- School of Medicine, The University of Western Australia, Crawley, WA, Australia; Harry Perkins Institute of Medical Research, Murdoch, WA, Australia
| | - Mazyar Ghaffari
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Jiao Jiao Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW, Australia
| | - Elham Hosseini-Beheshti
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia.
| |
Collapse
|
97
|
Quintanal-Villalonga A, Taniguchi H, Zhan YA, Hasan MM, Chavan SS, Meng F, Uddin F, Allaj V, Manoj P, Shah NS, Chan JM, Ciampricotti M, Chow A, Offin M, Ray-Kirton J, Egger JD, Bhanot UK, Linkov I, Asher M, Roehrl MH, Ventura K, Qiu J, de Stanchina E, Chang JC, Rekhtman N, Houck-Loomis B, Koche RP, Yu HA, Sen T, Rudin CM. Comprehensive molecular characterization of lung tumors implicates AKT and MYC signaling in adenocarcinoma to squamous cell transdifferentiation. J Hematol Oncol 2021; 14:170. [PMID: 34656143 PMCID: PMC8520275 DOI: 10.1186/s13045-021-01186-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/04/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Lineage plasticity, the ability to transdifferentiate among distinct phenotypic identities, facilitates therapeutic resistance in cancer. In lung adenocarcinomas (LUADs), this phenomenon includes small cell and squamous cell (LUSC) histologic transformation in the context of acquired resistance to targeted inhibition of driver mutations. LUAD-to-LUSC transdifferentiation, occurring in up to 9% of EGFR-mutant patients relapsed on osimertinib, is associated with notably poor prognosis. We hypothesized that multi-parameter profiling of the components of mixed histology (LUAD/LUSC) tumors could provide insight into factors licensing lineage plasticity between these histologies. METHODS We performed genomic, epigenomics, transcriptomics and protein analyses of microdissected LUAD and LUSC components from mixed histology tumors, pre-/post-transformation tumors and reference non-transformed LUAD and LUSC samples. We validated our findings through genetic manipulation of preclinical models in vitro and in vivo and performed patient-derived xenograft (PDX) treatments to validate potential therapeutic targets in a LUAD PDX model acquiring LUSC features after osimertinib treatment. RESULTS Our data suggest that LUSC transdifferentiation is primarily driven by transcriptional reprogramming rather than mutational events. We observed consistent relative upregulation of PI3K/AKT, MYC and PRC2 pathway genes. Concurrent activation of PI3K/AKT and MYC induced squamous features in EGFR-mutant LUAD preclinical models. Pharmacologic inhibition of EZH1/2 in combination with osimertinib prevented relapse with squamous-features in an EGFR-mutant patient-derived xenograft model, and inhibition of EZH1/2 or PI3K/AKT signaling re-sensitized resistant squamous-like tumors to osimertinib. CONCLUSIONS Our findings provide the first comprehensive molecular characterization of LUSC transdifferentiation, suggesting putative drivers and potential therapeutic targets to constrain or prevent lineage plasticity.
Collapse
Affiliation(s)
- Alvaro Quintanal-Villalonga
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, 408 East 69th Street, ZRC-1731, New York, NY, 10021, USA.
| | - Hirokazu Taniguchi
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, 408 East 69th Street, ZRC-1731, New York, NY, 10021, USA
| | - Yingqian A Zhan
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Maysun M Hasan
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Shweta S Chavan
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fanli Meng
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fathema Uddin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, 408 East 69th Street, ZRC-1731, New York, NY, 10021, USA
| | - Viola Allaj
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, 408 East 69th Street, ZRC-1731, New York, NY, 10021, USA
| | - Parvathy Manoj
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, 408 East 69th Street, ZRC-1731, New York, NY, 10021, USA
| | - Nisargbhai S Shah
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, 408 East 69th Street, ZRC-1731, New York, NY, 10021, USA
| | - Joseph M Chan
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, 408 East 69th Street, ZRC-1731, New York, NY, 10021, USA
- Program for Computational and Systems Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Metamia Ciampricotti
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, 408 East 69th Street, ZRC-1731, New York, NY, 10021, USA
| | - Andrew Chow
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, 408 East 69th Street, ZRC-1731, New York, NY, 10021, USA
| | - Michael Offin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, 408 East 69th Street, ZRC-1731, New York, NY, 10021, USA
| | - Jordana Ray-Kirton
- Precision Pathology Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jacklynn D Egger
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, 408 East 69th Street, ZRC-1731, New York, NY, 10021, USA
| | - Umesh K Bhanot
- Precision Pathology Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Irina Linkov
- Precision Pathology Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marina Asher
- Precision Pathology Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael H Roehrl
- Precision Pathology Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Katia Ventura
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Juan Qiu
- Antitumor Assessment Core, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Elisa de Stanchina
- Antitumor Assessment Core, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Jason C Chang
- Precision Pathology Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Natasha Rekhtman
- Precision Pathology Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Brian Houck-Loomis
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Richard P Koche
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Helena A Yu
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, 408 East 69th Street, ZRC-1731, New York, NY, 10021, USA
- Weill Cornell Medical College, 1275 York Avenue, New York, NY, 10065, USA
| | - Triparna Sen
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, 408 East 69th Street, ZRC-1731, New York, NY, 10021, USA.
- Weill Cornell Medical College, 1275 York Avenue, New York, NY, 10065, USA.
| | - Charles M Rudin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, 408 East 69th Street, ZRC-1731, New York, NY, 10021, USA.
- Weill Cornell Medical College, 1275 York Avenue, New York, NY, 10065, USA.
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
98
|
Quan Y, Zhang X, Butler W, Du Z, Wang M, Liu Y, Ping H. The role of N-cadherin/c-Jun/NDRG1 axis in the progression of prostate cancer. Int J Biol Sci 2021; 17:3288-3304. [PMID: 34512147 PMCID: PMC8416735 DOI: 10.7150/ijbs.63300] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 07/06/2021] [Indexed: 02/03/2023] Open
Abstract
The dysregulation of androgen receptor (AR) signaling is a critical event in the progression of prostate cancer (PCa) and hormone therapy consisting of androgen deprivation (ADT) or AR inhibition is therefore used to treat advanced cases. It is known that N-cadherin becomes upregulated following ADT and can directly induce PCa transformation to the castration-resistant stage (CRPC). However, the relationship between AR and N-cadherin is unclear and may promote better understanding of CRPC pathogenesis and progression. Here, we demonstrate a new axis of N-cadherin/c-Jun/N-myc downstream regulated gene 1 (NDRG1) that N-cadherin promotes c-Jun expression and suppresses NDRG1 to promote invasion and migration of PCa cells through epithelial to mesenchymal transition (EMT). Targeting N-cadherin in combination with enzalutamide (ENZ) treatment synergistically suppressed PC3 cell proliferation in vivo and in vitro. Further studies showed that compared to lower Gleason score (GS) (GS < 7) cases, high GS (GS > 7) cases exhibited elevated N-cadherin expression and reduced NDRG1 expression, corroborating our in vitro observations. We further demonstrate that c-Jun, AR, and DNA methyltransferase-1 (DNMT1) form a complex in the 12-O-tetradecanoyl phorbol-13-acetate (TPA) response elements (TREs) region of the NDRG1 promoter, which suppresses NDRG1 transcription through DNA hypermethylation. In conclusion, we demonstrate an underlying mechanism for how N-cadherin collaborates with AR and NDRG1 to promote CRPC progression. Controlling N-cadherin/c-Jun/NDRG1 axis may help to overcome resistance to commonly used hormone therapy to improve long-term patient outcomes.
Collapse
Affiliation(s)
- Yongjun Quan
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Xiaodong Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - William Butler
- Department of Pathology, Duke University School of Medicine, Durham NC 27710, USA
| | - Zhen Du
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Mingdong Wang
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Yuexin Liu
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Hao Ping
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| |
Collapse
|
99
|
Park SH, Fong KW, Mong E, Martin MC, Schiltz GE, Yu J. Going beyond Polycomb: EZH2 functions in prostate cancer. Oncogene 2021; 40:5788-5798. [PMID: 34349243 PMCID: PMC8487936 DOI: 10.1038/s41388-021-01982-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 02/07/2023]
Abstract
The Polycomb group (PcG) protein Enhancer of Zeste Homolog 2 (EZH2) is one of the three core subunits of the Polycomb Repressive Complex 2 (PRC2). It harbors histone methyltransferase activity (MTase) that specifically catalyze histone 3 lysine 27 (H3K27) methylation on target gene promoters. As such, PRC2 are epigenetic silencers that play important roles in cellular identity and embryonic stem cell maintenance. In the past two decades, mounting evidence supports EZH2 mutations and/or over-expression in a wide array of hematological cancers and solid tumors, including prostate cancer. Further, EZH2 is among the most upregulated genes in neuroendocrine prostate cancers, which become abundant due to the clinical use of high-affinity androgen receptor pathway inhibitors. While numerous studies have reported epigenetic functions of EZH2 that inhibit tumor suppressor genes and promote tumorigenesis, discordance between EZH2 and H3K27 methylation has been reported. Further, enzymatic EZH2 inhibitors have shown limited efficacy in prostate cancer, warranting a more comprehensive understanding of EZH2 functions. Here we first review how canonical functions of EZH2 as a histone MTase are regulated and describe the various mechanisms of PRC2 recruitment to the chromatin. We further outline non-histone substrates of EZH2 and discuss post-translational modifications to EZH2 itself that may affect substrate preference. Lastly, we summarize non-canonical functions of EZH2, beyond its MTase activity and/or PRC2, as a transcriptional cofactor and discuss prospects of its therapeutic targeting in prostate cancer.
Collapse
Affiliation(s)
- Su H Park
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ka-Wing Fong
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - Ezinne Mong
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - M Cynthia Martin
- Department of Chemistry, Northwestern University, Evanston, IL, USA
| | - Gary E Schiltz
- Department of Chemistry, Northwestern University, Evanston, IL, USA
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Jindan Yu
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA.
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
100
|
Berchuck JE, Viscuse PV, Beltran H, Aparicio A. Clinical considerations for the management of androgen indifferent prostate cancer. Prostate Cancer Prostatic Dis 2021; 24:623-637. [PMID: 33568748 PMCID: PMC8353003 DOI: 10.1038/s41391-021-00332-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/16/2020] [Accepted: 01/20/2021] [Indexed: 02/01/2023]
Abstract
BACKGROUND Many systemic therapies for advanced prostate cancer work by disrupting androgen receptor signaling. Androgen indifferent prostate cancer (AIPC) variants, including aggressive variant prostate cancer (AVPC), neuroendocrine prostate cancer (NEPC), and double-negative prostate cancer (DNPC), are increasingly common and often overlapping resistance phenotypes following treatment with androgen receptor signaling inhibitors in men with metastatic castration-resistant prostate cancer and are associated with poor outcomes. Understanding the underlying biology and identifying effective therapies for AIPC is paramount for improving survival for men with prostate cancer. METHODS In this review, we summarize the current knowledge on AIPC variants, including our current understanding of the clinical, morphologic, and molecular features as well as current therapeutic approaches. We also explore emerging therapies and biomarkers aimed at improving outcomes for men with AIPC. RESULTS AND CONCLUSIONS Establishing consensus definitions, developing novel biomarkers for early and accurate detection, further characterization of molecular drivers of each phenotype, and developing effective therapies will be critical to improving outcomes for men with AIPC. Significant progress has been made toward defining the clinical and molecular characteristics of AVPC, NEPC, and DNPC. Novel diagnostic approaches, including cell-free DNA, circulating tumor cells, and molecular imaging are promising tools for detecting AIPC in clinical practice. Building on previous treatment advances, several clinical trials are underway evaluating novel therapeutic approaches in patients with AIPC informed by an understanding of variant-specific biology. In this review, we discuss how these recent and ongoing studies will help to improve diagnosis, prognosis, and therapy for men with AIPC.
Collapse
Affiliation(s)
- Jacob E Berchuck
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Paul V Viscuse
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Ana Aparicio
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|