1
|
Danielli L, Tassinari E, Marchetti A, Rosellini M, Mollica V, Cheng L, Massari F. Current androgen receptor antagonists under investigation for resistant prostate cancer: progress and challenges. Expert Rev Anticancer Ther 2025; 25:457-470. [PMID: 40089934 DOI: 10.1080/14737140.2025.2481141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/27/2025] [Accepted: 03/14/2025] [Indexed: 03/17/2025]
Abstract
INTRODUCTION Prostate cancer represents a significant oncological challenge, with its natural history predominantly driven by androgen receptor (AR) signaling. The pivotal role of this pathway underscores the rationale for targeting AR activity in therapeutic strategies. However, the development of resistance mechanisms has highlighted the need for advanced therapies to address the complexity of the castration-resistant status. AREAS COVERED We analyzed the evolving role of second-generation androgen receptor signaling inhibitors (ARSIs) in the management of non-metastatic and metastatic castration-resistant prostate cancer, we critically examine emerging combination strategies involving ARSIs, novel agents targeting resistance pathways, and the mechanisms underlying treatment resistance. The review also provides insights into future directions for enhancing outcomes. PubMed literature research using keywords related to castration-resistant prostate cancer and its treatments was performed, including the most relevant trials and reviews. EXPERT OPINION ARSIs have revolutionized the management of prostate cancer, providing substantial clinical benefits and representing the cornerstone of current treatment paradigms. However, key challenges remain, including determining optimal treatment sequencing, overcoming resistance mechanisms, and tailoring therapies to specific molecular subtypes. Biomarker-driven approaches are critical for refining patient selection and improving therapeutic outcomes. Ongoing trials investigating novel hormonal-axis-directed agents and innovative combination therapies aim to expand the arsenal of effective treatment.
Collapse
Affiliation(s)
- Linda Danielli
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Elisa Tassinari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Andrea Marchetti
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Matteo Rosellini
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Department of Surgery (Urology), Brown University Warren Alpert Medical School, The Legorreta Cancer Center at Brown University, and Brown University Health, Providence, RI, USA
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| |
Collapse
|
2
|
Pandey SK, Sabharwal U, Tripathi S, Mishra A, Yadav N, Dwivedi-Agnihotri H. Androgen Signaling in Prostate Cancer: When a Friend Turns Foe. Endocr Metab Immune Disord Drug Targets 2025; 25:37-56. [PMID: 38831575 DOI: 10.2174/0118715303313528240523101940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/17/2024] [Accepted: 05/02/2024] [Indexed: 06/05/2024]
Abstract
Androgen (AR) signaling is the main signaling for the development of the prostate and its normal functioning. AR is highly specific for testosterone and dihydrotestosterone, significantly contributing to prostate development, physiology, and cancer. All these receptors have emerged as crucial therapeutic targets for PCa. In the year 1966, the Noble prize was awarded to Huggins and Hodge for their groundbreaking discovery of AR. As it is a pioneer transcription factor, it belongs to the steroid hormone receptor family and consists of domains, including DNA binding domain (DBD), hormone response elements (HRE), C-terminal ligand binding domain (LBD), and N-terminal regulatory domains. Structural variations in AR, such as AR gene amplification, LBD mutations, alternative splicing of exons, hypermethylation of AR, and co- regulators, are major contributors to PCa. It's signaling is crucial for the development and functioning of the prostate gland, with the AR being the key player. The specificity of AR for testosterone and dihydrotestosterone is important in prostate physiology. However, when it is dysregulated, AR contributes significantly to PCa. However, the structural variations in AR, such as gene amplification, mutations, alternative splicing, and epigenetic modifications, drive the PCa progression. Therefore, understanding AR function and dysregulation is essential for developing effective therapeutic strategies. Thus, the aim of this review was to examine how AR was initially pivotal for prostate development and how it turned out to show both positive and detrimental implications for the prostate.
Collapse
Affiliation(s)
- Swaroop Kumar Pandey
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura, 281406, India
| | - Usha Sabharwal
- P. G. Department of Biosciences, Centre of Advanced Studies, Satellite Campus, Sardar Patel Maidan, 388120, Gujarat, India
| | - Swati Tripathi
- Section of Electron Microscopy, Supportive Centre for Brain Research, National Institute for Physiological Sciences (NIPS) Okazaki, 444-8787, Japan
| | - Anuja Mishra
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura, 281406, India
| | - Neha Yadav
- Department of Biophysics, University of Delhi, South Campus, New Delhi, 110021, India
| | | |
Collapse
|
3
|
Kim JH, Bae GH, Jung J, Noh TI. Secondary Cancer after Androgen Deprivation Therapy in Prostate Cancer: A Nationwide Study. World J Mens Health 2025; 43:123-133. [PMID: 38606859 PMCID: PMC11704168 DOI: 10.5534/wjmh.230237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/15/2023] [Accepted: 12/05/2023] [Indexed: 04/13/2024] Open
Abstract
PURPOSE Androgen signaling is associated with various secondary cancer, which could be promising for potential treatment using androgen deprivation therapy (ADT). This study investigated whether ADT use was associated with secondary cancers other than prostate cancer in a nationwide population-based cohort. MATERIALS AND METHODS A total, 278,434 men with newly diagnosed prostate cancer between January 1, 2002 and December 31, 2017 were identified. After applying the exclusion criteria, 170,416 men were enrolled. The study cohort was divided into ADT and non-ADT groups by individual matching followed by propensity score matching (PSM). Study outcomes were incidence of all male cancers. Cox proportional hazard regression models were used to estimate adjusted hazard ratios (HRs) and 95% confidence intervals (CIs) of events. RESULTS During a median follow-up of 4.5 years, a total of 11,059 deaths (6,329 in the ADT group and 4,730 in the non-ADT group) after PSM were found. After PSM, the overall all-cause of secondary cancer incidence risk of the ADT group was higher than that of the non-ADT group (HR: 1.312, 95% CI: 1.23-1.36; adjusted HR: 1.344, 95% CI: 1.29-1.40). The ADT group showed higher risk of overall brain and other central nervous system (CNS) cancer-specific incidence than the non-ADT group (adjusted HR: 1.648, 95% CI: 1.21-2.24). The ADT group showed lower risks of overall cancer-specific incidence for stomach, colon/rectum, liver/inflammatory bowel disease (IBD), gall bladder/extrahepatic bile duct, lung, bladder, and kidney cancers than the non-ADT group. When the duration of ADT was more than 2 years of ADT, the ADT group showed higher risk of cancer-specific incidence for brain and other CNS cancers but lower risk of cancer-specific incidence for liver/IBD and lung cancers than the non-ADT group. CONCLUSIONS This study demonstrates that ADT could affect cancer-specific incidence for various cancers.
Collapse
Affiliation(s)
- Jae Heon Kim
- Department of Urology, Soonchunhyang University Seoul Hospital, Soonchunhyang University College of Medicine, Seoul, Korea
| | - Gi Hwan Bae
- Artificial Intelligence and Big-Data Convergence Center, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - Jaehun Jung
- Artificial Intelligence and Big-Data Convergence Center, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
- Department of Preventive Medicine, Gachon University College of Medicine, Incheon, Korea.
| | - Tae Il Noh
- Department of Urology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea.
| |
Collapse
|
4
|
Aswani BS, Hegde M, Vishwa R, Alqahtani MS, Abbas M, Almubarak HA, Sethi G, Kunnumakkara AB. Tackling exosome and nuclear receptor interaction: an emerging paradigm in the treatment of chronic diseases. Mil Med Res 2024; 11:67. [PMID: 39327610 PMCID: PMC11426102 DOI: 10.1186/s40779-024-00564-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 08/06/2024] [Indexed: 09/28/2024] Open
Abstract
Nuclear receptors (NRs) function as crucial transcription factors in orchestrating essential functions within the realms of development, host defense, and homeostasis of body. NRs have garnered increased attention due to their potential as therapeutic targets, with drugs directed at NRs demonstrating significant efficacy in impeding chronic disease progression. Consequently, these pharmacological agents hold promise for the treatment and management of various diseases. Accumulating evidence emphasizes the regulatory role of exosome-derived microRNAs (miRNAs) in chronic inflammation, disease progression, and therapy resistance, primarily by modulating transcription factors, particularly NRs. By exploiting inflammatory pathways such as protein kinase B (Akt)/mammalian target of rapamycin (mTOR), nuclear factor kappa-B (NF-κB), signal transducer and activator of transcription 3 (STAT3), and Wnt/β-catenin signaling, exosomes and NRs play a pivotal role in the panorama of development, physiology, and pathology. The internalization of exosomes modulates NRs and initiates diverse autocrine or paracrine signaling cascades, influencing various processes in recipient cells such as survival, proliferation, differentiation, metabolism, and cellular defense mechanisms. This comprehensive review meticulously examines the involvement of exosome-mediated NR regulation in the pathogenesis of chronic ailments, including atherosclerosis, cancer, diabetes, liver diseases, and respiratory conditions. Additionally, it elucidates the molecular intricacies of exosome-mediated communication between host and recipient cells via NRs, leading to immunomodulation. Furthermore, it outlines the implications of exosome-modulated NR pathways in the prophylaxis of chronic inflammation, delineates current limitations, and provides insights into future perspectives. This review also presents existing evidence on the role of exosomes and their components in the emergence of therapeutic resistance.
Collapse
Affiliation(s)
- Babu Santha Aswani
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Ravichandran Vishwa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, 61421, Abha, Saudi Arabia
- BioImaging Unit, Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester, LE1 7RH, UK
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, 61421, Abha, Saudi Arabia
| | - Hassan Ali Almubarak
- Division of Radiology, Department of Medicine, College of Medicine and Surgery, King Khalid University, 61421, Abha, Saudi Arabia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117699, Singapore.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India.
| |
Collapse
|
5
|
Huang J, Ali T, Feldman DM, Theise ND. Androgen-Induced, β-Catenin-Activated Hepatocellular Adenomatosis with Spontaneous External Rupture. Diagnostics (Basel) 2024; 14:1473. [PMID: 39061609 PMCID: PMC11276095 DOI: 10.3390/diagnostics14141473] [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: 05/07/2024] [Revised: 05/29/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
Androgens have long been recognized as oncogenic agents. They can induce both benign and malignant hepatocellular neoplasms, including hepatocellular adenoma (HCA) and hepatocellular carcinoma, though the underlying mechanisms remain unclear. Androgen-induced liver tumors are most often solitary and clinically silent. Herein, we reported an androgen-induced HCA complicated by spontaneous rupture. The patient was a 24-year-old male presenting with fatigue, diminished libido, radiology-diagnosed hepatocellular adenomatosis for 3 years, and sudden-onset, severe, sharp, constant abdominal pain for one day. He used Aveed (testosterone undecanoate injection) from age 17 and completely stopped one year before his presentation. A physical exam showed touch pain and voluntary guarding in the right upper quadrant of the abdomen. An abdominal CT angiogram demonstrated multiple probable HCAs, with active hemorrhage of the largest one (6.6 × 6.2 × 5.1 cm) accompanied by large-volume hemoperitoneum. After being stabilized by a massive transfusion protocol and interventional embolization, he underwent a percutaneous liver core biopsy. The biopsy specimen displayed atypical hepatocytes forming dense cords and pseudoglands. The lesional cells diffusely stained β-catenin in nuclei and glutamine synthetase in cytoplasm. Compared to normal hepatocytes from control tissue, the tumor cells were positive for nuclear AR (androgen receptor) expression but had no increased EZH2 (Enhancer of Zeste 2 Polycomb Repressive Complex 2 Subunit) protein expression. The case indicated that androgen-induced hepatocellular neoplasms should be included in the differential diagnosis of acute abdomen.
Collapse
Affiliation(s)
- Jialing Huang
- Department of Pathology, Geisinger Medical Center, Geisinger Commonwealth School of Medicine, 100 N. Academy Ave, Danville, PA 17822, USA
| | - Towhid Ali
- Department of Radiology, Geisinger Medical Center, Geisinger Commonwealth School of Medicine, 100 N. Academy Ave, Danville, PA 17822, USA
| | - David M. Feldman
- Department of Gastroenterology and Hepatology, New York University Grossman School of Medicine, 550 1st Avenue, New York, NY 10016, USA
| | - Neil D. Theise
- Department of Pathology, NYU Langone Medical Center, New York University Grossman School of Medicine, 550 1st Avenue, New York, NY 10016, USA
| |
Collapse
|
6
|
Hu YM, Zhao F, Graff JN, Chen C, Zhao X, Thomas GV, Wu H, Kardosh A, Mills GB, Alumkal JJ, Moran AE, Xia Z. Androgen receptor activity inversely correlates with immune cell infiltration and immunotherapy response across multiple cancer lineages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.08.593181. [PMID: 38798471 PMCID: PMC11118439 DOI: 10.1101/2024.05.08.593181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
There is now increasing recognition of the important role of androgen receptor (AR) in modulating immune function. To gain a comprehensive understanding of the effects of AR activity on cancer immunity, we employed a computational approach to profile AR activity in 33 human tumor types using RNA-Seq datasets from The Cancer Genome Atlas. Our pan-cancer analysis revealed that the genes most negatively correlated with AR activity across cancers are involved in active immune system processes. Importantly, we observed a significant negative correlation between AR activity and IFNγ pathway activity at the pan-cancer level. Indeed, using a matched biopsy dataset from subjects with prostate cancer before and after AR-targeted treatment, we verified that inhibiting AR enriches immune cell abundances and is associated with higher IFNγ pathway activity. Furthermore, by analyzing immunotherapy datasets in multiple cancers, our results demonstrate that low AR activity was significantly associated with a favorable response to immunotherapy. Together, our data provide a comprehensive assessment of the relationship between AR signaling and tumor immunity.
Collapse
Affiliation(s)
- Ya-Mei Hu
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Faming Zhao
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Julie N. Graff
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- VA Portland Health Care System, Portland, OR, USA
| | - Canping Chen
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Xiyue Zhao
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - George V. Thomas
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Pathology & Laboratory Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Hui Wu
- Division of Biomaterial and Biomedical Sciences, Department of Oral Rehabilitation and Biosciences, Oregon Health & Science University, Portland, OR, USA
| | - Adel Kardosh
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Gordon B. Mills
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Joshi J. Alumkal
- Department of Internal Medicine, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Amy E. Moran
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Zheng Xia
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Center for Biomedical Data Science, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
7
|
Hoffmann H, Ebensperger M, Schönsiegel A, Hamza H, Koch-Heier J, Schreiber A, Ludwig S, Schindler M, Planz O. Influenza A virus replication has a stronger dependency on Raf/MEK/ERK signaling pathway activity than SARS-CoV-2. Front Cell Infect Microbiol 2023; 13:1264983. [PMID: 37965261 PMCID: PMC10641236 DOI: 10.3389/fcimb.2023.1264983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 10/10/2023] [Indexed: 11/16/2023] Open
Abstract
The recent COVID-19 pandemic again highlighted the urgent need for broad-spectrum antivirals, both for therapeutic use in acute viral infection and for pandemic preparedness in general. The targeting of host cell factors hijacked by viruses during their replication cycle presents one possible strategy for development of broad-spectrum antivirals. By inhibiting the Raf/MEK/ERK signaling pathway, a central kinase cascade of eukaryotic cells, which is being exploited by numerous viruses of different virus phyla, the small-molecule MEK inhibitor zapnometinib has the potential to address this need. We here performed a side-by-side comparison of the antiviral efficacy of zapnometinib against IAV and SARS-CoV-2 to determine the concentration leading to 50% of its effect on the virus (EC50) and the concentration leading to 50% reduction of ERK phosphorylation (IC50) in a comparable manner, using the same experimental conditions. Our results show that the EC50 value and IC50 value of zapnometinib are indeed lower for IAV compared to SARS-CoV-2 using one representative strain for each. The results suggest that IAV's replication has a stronger dependency on an active Raf/MEK/ERK pathway and, thus, that IAV is more susceptible to treatment with zapnometinib than SARS-CoV-2. With zapnometinib's favorable outcome in a recent phase II clinical trial in hospitalized COVID-19 patients, the present results are even more promising for an upcoming phase II clinical trial in severe influenza virus infection.
Collapse
Affiliation(s)
- Helen Hoffmann
- Department of Immunology, Interfaculty Institute for Cell Biology, Eberhard Karls Universitaet Tuebingen, Tuebingen, Germany
- Atriva Therapeutics GmbH, Tuebingen, Germany
| | | | - Annika Schönsiegel
- Department of Immunology, Interfaculty Institute for Cell Biology, Eberhard Karls Universitaet Tuebingen, Tuebingen, Germany
- Atriva Therapeutics GmbH, Tuebingen, Germany
| | - Hazem Hamza
- Department of Immunology, Interfaculty Institute for Cell Biology, Eberhard Karls Universitaet Tuebingen, Tuebingen, Germany
- Virology Laboratory, Environmental Research Division, National Research Centre, Giza, Egypt
| | - Julia Koch-Heier
- Department of Immunology, Interfaculty Institute for Cell Biology, Eberhard Karls Universitaet Tuebingen, Tuebingen, Germany
- Atriva Therapeutics GmbH, Tuebingen, Germany
| | - André Schreiber
- Institute of Virology (IVM), Westfaelische Wilhelms Universitaet, Muenster, Muenster, Germany
| | - Stephan Ludwig
- Institute of Virology (IVM), Westfaelische Wilhelms Universitaet, Muenster, Muenster, Germany
| | - Michael Schindler
- Department of Molecular Virology, Institute for Medical Virology and Epidemiology of Viral Disease, University Hospital Tuebingen, Tuebingen, Germany
| | - Oliver Planz
- Department of Immunology, Interfaculty Institute for Cell Biology, Eberhard Karls Universitaet Tuebingen, Tuebingen, Germany
| |
Collapse
|
8
|
Nuvola G, Santoni M, Rizzo M, Rosellini M, Mollica V, Rizzo A, Marchetti A, Battelli N, Massari F. Adapting to hormone-therapy resistance for adopting the right therapeutic strategy in advanced prostate cancer. Expert Rev Anticancer Ther 2023; 23:593-600. [PMID: 37185042 DOI: 10.1080/14737140.2023.2207827] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
INTRODUCTION The androgen/androgen receptor (AR) axis represents a key driver of treatment resistance in prostate cancer (PCa) patients receiving androgen deprivation therapy (ADT) and targeted agents, and a deeper comprehension of resistance mechanisms is fundamental to adopt effective therapeutic strategies. AREAS COVERED We review the mechanisms of primary or secondary resistance to hormone therapy (HT) in PCa, especially focusing on available data and emerging evidence. EXPERT OPINION First- and second-generation HT resistance has been associated with several AR-dependent and AR-independent mechanisms, ranging from the amplification of the AR gene locus to somatic AR mutations and the intratumoral synthesis of androgens from adrenal steroids and cholesterol. As reported in the current review, the development of novel and effective treatments is needed to personalize anticancer therapies in this setting and to finally improve clinical outcomes in patients with HT resistant disease.
Collapse
Affiliation(s)
- Giacomo Nuvola
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni - 15, Bologna, Italia
| | - Matteo Santoni
- Oncology Unit, Macerata Hospital, via Santa Lucia 2, 62100, Macerata, Italy
| | - Mimma Rizzo
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, Piazza G. Cesare 11, 70124, Bari, Italy
| | - Matteo Rosellini
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni - 15, Bologna, Italia
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni - 15, Bologna, Italia
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Alessandro Rizzo
- Struttura Semplice Dipartimentale di Oncologia Medica per la Presa in Carico Globale del Paziente Oncologico "Don Tonino Bello", Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Istituto Tumori Giovanni Paolo II-Bari, 70124 Bari, Italy
| | - Andrea Marchetti
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni - 15, Bologna, Italia
| | - Nicola Battelli
- Oncology Unit, Macerata Hospital, via Santa Lucia 2, 62100, Macerata, Italy
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni - 15, Bologna, Italia
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| |
Collapse
|
9
|
Steiner I, Flores-Tellez TDNJ, Mevel R, Ali A, Wang P, Schofield P, Behan C, Forsythe N, Ashton G, Taylor C, Mills IG, Oliveira P, McDade SS, Zaiss DM, Choudhury A, Lacaud G, Baena E. Autocrine activation of MAPK signaling mediates intrinsic tolerance to androgen deprivation in LY6D prostate cancer cells. Cell Rep 2023; 42:112377. [PMID: 37060563 DOI: 10.1016/j.celrep.2023.112377] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/12/2022] [Accepted: 03/23/2023] [Indexed: 04/16/2023] Open
Abstract
The emergence of castration-resistant prostate cancer remains an area of unmet clinical need. We recently identified a subpopulation of normal prostate progenitor cells, characterized by an intrinsic resistance to androgen deprivation and expression of LY6D. We here demonstrate that conditional deletion of PTEN in the murine prostate epithelium causes an expansion of transformed LY6D+ progenitor cells without impairing stem cell properties. Transcriptomic analyses of LY6D+ luminal cells identified an autocrine positive feedback loop, based on the secretion of amphiregulin (AREG)-mediated activation of mitogen-activated protein kinase (MAPK) signaling, increasing cellular fitness and organoid formation. Pharmacological interference with this pathway overcomes the castration-resistant properties of LY6D+ cells with a suppression of organoid formation and loss of LY6D+ cells in vivo. Notably, LY6D+ tumor cells are enriched in high-grade and androgen-resistant prostate cancer, providing clinical evidence for their contribution to advanced disease. Our data indicate that early interference with MAPK inhibitors can prevent progression of castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Ivana Steiner
- Prostate Oncobiology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK
| | - Teresita Del N J Flores-Tellez
- Prostate Oncobiology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK
| | - Renaud Mevel
- Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK
| | - Amin Ali
- Prostate Oncobiology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK; Belfast-Manchester Movember Centre of Excellence, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK
| | - Pengbo Wang
- Prostate Oncobiology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK
| | - Pieta Schofield
- Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK
| | - Caron Behan
- Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK
| | - Nicholas Forsythe
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, BT9 7BL Northern Ireland, UK; Belfast-Manchester Movember Centre of Excellence, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK
| | - Garry Ashton
- Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK
| | - Catherine Taylor
- The Christie NHS Foundation Trust, Manchester Academic Health Sciences Centre, M20 4BX Manchester, UK
| | - Ian G Mills
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, BT9 7BL Northern Ireland, UK; Belfast-Manchester Movember Centre of Excellence, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK; Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, OX3 9DU Oxford, UK; Department of Clinical Sciences and Centre for Cancer Biomarkers, University of Bergen, 7804 Bergen, Norway
| | - Pedro Oliveira
- Department of Pathology, The Christie NHS Foundation Trust, M20 4BX Manchester, UK
| | - Simon S McDade
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, BT9 7BL Northern Ireland, UK; Belfast-Manchester Movember Centre of Excellence, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK
| | - Dietmar M Zaiss
- Department of Immune Medicine, University Regensburg, Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, and Leibniz Institute for Immunotherapy (LIT), 93053 Regensburg, Germany
| | - Ananya Choudhury
- The Christie NHS Foundation Trust, Manchester Academic Health Sciences Centre, M20 4BX Manchester, UK; The University of Manchester, Manchester Cancer Research Centre, M20 4BX Manchester, UK; Belfast-Manchester Movember Centre of Excellence, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK
| | - Georges Lacaud
- Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK
| | - Esther Baena
- Prostate Oncobiology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK; Belfast-Manchester Movember Centre of Excellence, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK.
| |
Collapse
|
10
|
Dahiya V, Bagchi G. Non-canonical androgen signaling pathways and implications in prostate cancer. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119357. [PMID: 36100060 DOI: 10.1016/j.bbamcr.2022.119357] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/11/2022] [Accepted: 09/01/2022] [Indexed: 06/15/2023]
Abstract
Androgen signaling is a critical determinant of timely and proper development of all male organs including the prostate. Maturation of prostate and its neoplastic transformation is intricately associated with accurate androgen signaling. Ablation of androgen has therefore been the primary treatment mechanism of Prostate cancer (PCa) patients for several decades. Upon removal, the tumor recedes for a while, yet it reappears soon, in an androgen independent state, untreatable by current therapeutic regimens. Studies reveal that apart from the classical androgen signaling pathway known and targeted for almost a century, there exist several non-canonical pathways, with marked impact on classical androgen signaling and PCa growth. These include non-genomic signaling by androgens via alternate membrane GPCRs, signaling by non-androgens that ultimately impact the androgen signaling pathway, or an integration of non-genomic and genomic response as seen in case of protein kinase A activation. Accurate understanding of these various non-canonical androgen signaling pathways and their influence on the typical androgen signaling pathway can help design important interventions for PCa patients. This review analyses in detail the various non-classical androgen signaling pathways and their impact, if any, on classical mode of androgen action and PCa.
Collapse
Affiliation(s)
- Versha Dahiya
- Amity Institute of Biotechnology, Amity University Haryana, India, 122413
| | - Gargi Bagchi
- Amity Institute of Biotechnology, Amity University Haryana, India, 122413.
| |
Collapse
|
11
|
Edachery S, Patil P, Mohan R, Aradhya B, Shetty J, Kabekkodu SP, Santra MK, Gonchigar SJ, Shetty P. Loss of miR-936 leads to acquisition of androgen-independent metastatic phenotype in prostate cancer. Sci Rep 2022; 12:17070. [PMID: 36224238 PMCID: PMC9556567 DOI: 10.1038/s41598-022-20777-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/19/2022] [Indexed: 12/30/2022] Open
Abstract
Prostate cancer (PCa) progresses from a hormone-sensitive, androgen-dependent to a hormone-refractory, androgen-independent metastatic phenotype. Among the many genes implicated, ANXA2, a calcium-dependent phospholipid binding protein, has been found to have a critical role in the progression of PCa into more invasive metastatic phenotype. However, the molecular mechanisms underlying the absence of ANXA2 in early PCa and its recurrence in advanced stage are yet unknown. Moreover, recent studies have observed the deregulation of microRNAs (miRNAs) are involved in the development and progression of PCa. In this study, we found the down-regulation of miR-936 in metastatic PCa wherein its target ANXA2 was overexpressed. Subsequently, it has been shown that the downregulation of miRNA biogenesis by siRNA treatment in ANXA2-null LNCaP cells could induce the expression of ANXA2, indicating the miRNA mediated regulation of ANXA2 expression. Additionally, we demonstrate that miR-936 regulates ANXA2 expression by direct interaction at coding as well as 3'UTR region of ANXA2 mRNA by luciferase reporter assay. Furthermore, the overexpression of miR-936 suppresses the cell proliferation, cell cycle progression, cell migration, and invasion abilities of metastatic PCa PC-3 cells in vitro and tumor forming ability in vivo. These results indicate that miR-936 have tumor suppressor properties by regulating the over expression of ANXA2 in hormone-independent metastatic PCa. Moreover, our results suggest that this tumor suppressor miR-936 could be developed as a targeted therapeutic molecule for metastatic PCa control and to improve the prognosis in PCa patients.
Collapse
Affiliation(s)
- Sarathkumar Edachery
- Department of Biochemistry, Kuvempu University, Shankaraghatta, Karnataka, 577451, India
- Division of Proteomics and Cancer Biology, Nitte University Center for Science Education and Research, Nitte (Deemed to be University), Mangaluru, Karnataka, 575018, India
| | - Prakash Patil
- Central Research Laboratory, K S Hegde Medical Academy, Nitte (Deemed to be University), Deralakatte, Mangaluru, 575018, India
| | - Rajashekar Mohan
- Department of Surgery, All India Institute of Medical Sciences, Mangalagiri, Andhra Pradesh, 522503, India
| | | | - Jayaprakash Shetty
- Department of Pathology, K S Hegde Medical Academy, Nitte (Deemed to be University), Deralakatte, Mangaluru, 575018, India
| | | | | | | | - Praveenkumar Shetty
- Central Research Laboratory, K S Hegde Medical Academy, Nitte (Deemed to be University), Deralakatte, Mangaluru, 575018, India.
- Department of Biochemistry, K S Hegde Medical Academy, Nitte (Deemed to be University), Deralakatte, Mangaluru, 575018, India.
| |
Collapse
|
12
|
Luthold C, Hallal T, Labbé DP, Bordeleau F. The Extracellular Matrix Stiffening: A Trigger of Prostate Cancer Progression and Castration Resistance? Cancers (Basel) 2022; 14:cancers14122887. [PMID: 35740556 PMCID: PMC9221142 DOI: 10.3390/cancers14122887] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 02/06/2023] Open
Abstract
Despite advancements made in diagnosis and treatment, prostate cancer remains the second most diagnosed cancer among men worldwide in 2020, and the first in North America and Europe. Patients with localized disease usually respond well to first-line treatments, however, up to 30% develop castration-resistant prostate cancer (CRPC), which is often metastatic, making this stage of the disease incurable and ultimately fatal. Over the last years, interest has grown into the extracellular matrix (ECM) stiffening as an important mediator of diseases, including cancers. While this process is increasingly well-characterized in breast cancer, a similar in-depth look at ECM stiffening remains lacking for prostate cancer. In this review, we scrutinize the current state of literature regarding ECM stiffening in prostate cancer and its potential association with disease progression and castration resistance.
Collapse
Affiliation(s)
- Carole Luthold
- Centre de Recherche sur le Cancer, Université Laval, Québec, QC G1R 3S3, Canada;
- Division of Oncology, Centre de Recherche du CHU de Québec-Université Laval, Hôtel-Dieu de Québec, Québec, QC G1R 3S3, Canada
| | - Tarek Hallal
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada;
| | - David P. Labbé
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada;
- Division of Urology, Department of Surgery, McGill University, Montréal, QC H4A 3J1, Canada
- Correspondence: (D.P.L.); (F.B.)
| | - François Bordeleau
- Centre de Recherche sur le Cancer, Université Laval, Québec, QC G1R 3S3, Canada;
- Division of Oncology, Centre de Recherche du CHU de Québec-Université Laval, Hôtel-Dieu de Québec, Québec, QC G1R 3S3, Canada
- Département de Biologie Moléculaire, Biochimie Médicale et Pathologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
- Correspondence: (D.P.L.); (F.B.)
| |
Collapse
|
13
|
Moreira-Silva F, Henrique R, Jerónimo C. From Therapy Resistance to Targeted Therapies in Prostate Cancer. Front Oncol 2022; 12:877379. [PMID: 35686097 PMCID: PMC9170957 DOI: 10.3389/fonc.2022.877379] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/25/2022] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer (PCa) is the second most common malignancy among men worldwide. Although early-stage disease is curable, advanced stage PCa is mostly incurable and eventually becomes resistant to standard therapeutic options. Different genetic and epigenetic alterations are associated with the development of therapy resistant PCa, with specific players being particularly involved in this process. Therefore, identification and targeting of these molecules with selective inhibitors might result in anti-tumoral effects. Herein, we describe the mechanisms underlying therapy resistance in PCa, focusing on the most relevant molecules, aiming to enlighten the current state of targeted therapies in PCa. We suggest that selective drug targeting, either alone or in combination with standard treatment options, might improve therapeutic sensitivity of resistant PCa. Moreover, an individualized analysis of tumor biology in each PCa patient might improve treatment selection and therapeutic response, enabling better disease management.
Collapse
Affiliation(s)
- Filipa Moreira-Silva
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (He-alth Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC), Porto, Portugal
| | - Rui Henrique
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (He-alth Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC), Porto, Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal.,Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences of the University of Porto (ICBAS-UP), Porto, Portugal
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (He-alth Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC), Porto, Portugal.,Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences of the University of Porto (ICBAS-UP), Porto, Portugal
| |
Collapse
|
14
|
Budi HS, Ahmad FN, Achmad H, Ansari MJ, Mikhailova MV, Suksatan W, Chupradit S, Shomali N, Marofi F. Human epidermal growth factor receptor 2 (HER2)-specific chimeric antigen receptor (CAR) for tumor immunotherapy; recent progress. Stem Cell Res Ther 2022; 13:40. [PMID: 35093187 PMCID: PMC8800342 DOI: 10.1186/s13287-022-02719-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 01/11/2022] [Indexed: 12/17/2022] Open
Abstract
Due to the overexpression or amplification of human epidermal growth factor receptor 2 (HER2) with poor prognosis in a myriad of human tumors, recent studies have focused on HER2-targeted therapies. Deregulation in HER2 signaling pathways is accompanied by sustained tumor cells growth concomitant with their migration and also tumor angiogenesis and metastasis by stimulation of proliferation of a network of blood vessels. A large number of studies have provided clear evidence that the emerging HER2-directed treatments could be the outcome of patients suffering from HER2 positive breast and also gastric/gastroesophageal cancers. Thanks to its great anti-tumor competence, immunotherapy using HER2-specific chimeric antigen receptor (CAR) expressing immune cell has recently attracted increasing attention. Human T cells and also natural killer (NK) cells can largely be found in the tumor microenvironment, mainly contributing to the tumor immune surveillance. Such properties make them perfect candidate for genetically modification to express constructed CARs. Herein, we will describe the potential targets of the HER2 signaling in tumor cells to clarify HER2-mediated tumorigenesis and also discuss recent findings respecting the HER2-specific CAR-expressing immune cells (CAR T and CAR NK cell) for the treatment of HER2-expressing tumors.
Collapse
Affiliation(s)
- Hendrik Setia Budi
- Department of Oral Biology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, 60132 Indonesia
| | | | - Harun Achmad
- Department of Pediatric Dentistry, Faculty of Dentistry, Hasanuddin University, Makassar, Indonesia
| | - Mohammad Javed Ansari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | | | - Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, 10210 Thailand
| | - Supat Chupradit
- Department of Occupational Therapy, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200 Thailand
| | - Navid Shomali
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faroogh Marofi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
15
|
The Metastasis Suppressor NDRG1 Directly Regulates Androgen Receptor Signaling in Prostate Cancer. J Biol Chem 2021; 297:101414. [PMID: 34785213 PMCID: PMC8668986 DOI: 10.1016/j.jbc.2021.101414] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 11/07/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022] Open
Abstract
N-myc-downregulated gene 1 (NDRG1) has potent anticancer effects and inhibits cell growth, survival, metastasis, and angiogenesis. Previous studies suggested that NDRG1 is linked to the androgen signaling network, but this mechanistic relationship is unclear. Considering the crucial role of the androgen receptor (AR) in prostate cancer (PCa) progression, here we examined for the first time the effect of NDRG1 on AR expression, activation, and downstream signaling in LNCaP, 22Rv1, and C4-2B PCa cell types. We demonstrate that NDRG1 effectively promotes interaction of AR with the chaperone HSP90, which in turn stabilizes the AR while decreasing its androgen-mediated activation. The expression of NDRG1 suppressed: (1) AR activation, as measured by p-ARSer213 and p-ARSer81; (2) expression of a major AR transcriptional target, prostate-specific antigen (PSA); and (3) AR transcriptional activity, probably via inhibiting the c-Jun-AR interaction by reducing c-Jun phosphorylation (p-c-JunSer63). NDRG1 was also demonstrated to inhibit multiple key molecules involved in androgen-dependent and -independent signaling (namely EGFR, HER2, HER3, PI3K, STAT3, and NF-κB), which promote the development of castration-resistant prostate cancer. We also identified the cysteine-rich secretory protein/antigen 5/pathogenesis related-1 (CAP) domain of NDRG1 as vital for inhibition of AR activity. Examining NDRG1 and p-NDRG1 in PCa patient specimens revealed a significant negative correlation between NDRG1 and PSA levels in prostatectomy patients that went on to develop metastasis. These results highlight a vital role for NDRG1 in androgen signaling and its potential as a key therapeutic target and biomarker in PCa.
Collapse
|
16
|
Zalcman N, Gutreiman M, Shahar T, Weller M, Lavon I. Androgen Receptor Activation in Glioblastoma Can Be Achieved by Ligand-Independent Signaling through EGFR-A Potential Therapeutic Target. Int J Mol Sci 2021; 22:10954. [PMID: 34681618 PMCID: PMC8535837 DOI: 10.3390/ijms222010954] [Citation(s) in RCA: 8] [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: 08/26/2021] [Revised: 09/30/2021] [Accepted: 10/07/2021] [Indexed: 12/05/2022] Open
Abstract
Androgen receptor (AR) is a ligand-mediated transcription factor that belongs to the superfamily of steroid receptors. AR is overexpressed in most glioblastomas and is a potential therapeutic target. In prostate and breast cancers, AR activation can be achieved also by a ligand-independent signaling through receptor tyrosine kinases such as epidermal growth factor receptor (EGFR). Considering its major role in glioblastoma, we explored whether EGFR is involved in AR signaling in this tumor. Analysis of mRNA expression in 28 glioblastoma samples with quantitative real-time reverse-transcription polymerase chain reaction revealed a positive and significant correlation between AR and EGFR mRNA expression levels (R = 0.47, p = 0.0092), which was validated by The Cancer Genome Atlas dataset (n = 671) analysis (R = 0.3, p = 0.00006). Using Western blotting and immunofluorescence staining, we showed that the transduced overexpression of EGFR or its variant EGFRvIII in the U87MG cells induced AR protein overexpression and nuclear translocation and Protein kinase B (AKT) S473 and AR S210/213 phosphorylation. The EGFR kinase inhibitor afatinib and the AKT inhibitor MK2206 reduced AR nuclear translocation. Afatinib diminished AKT phosphorylation at 30 min and 6 h in the EGFR- and EGFRvIII-overexpressing cells, respectively, and decreased AR phosphorylation in EGFR-overexpressing cells at 4 h. Afatinib or MK2206 combination therapy with the AR antagonist enzalutamide in the EGFR and EGFRvIII-overexpressing cells had synergistic efficacy. Our findings suggest that EGFR signaling is involved in AR activation in glioblastoma and buttresses the concept of combining an EGFR signaling inhibitor with AR antagonists as a potential glioblastoma treatment.
Collapse
Affiliation(s)
- Nomi Zalcman
- Molecular Neuro-Oncology Laboratory, Leslie and Michael Gaffin Center for Neuro-Oncology, Agnes Ginges Center for Human Neurogenetics, Neurology Department, Hadassah Hebrew University Medical Center, P.O. Box 12000, Jerusalem 91120, Israel; (N.Z.); (M.G.)
| | - Mijal Gutreiman
- Molecular Neuro-Oncology Laboratory, Leslie and Michael Gaffin Center for Neuro-Oncology, Agnes Ginges Center for Human Neurogenetics, Neurology Department, Hadassah Hebrew University Medical Center, P.O. Box 12000, Jerusalem 91120, Israel; (N.Z.); (M.G.)
| | - Tal Shahar
- The Laboratory for Molecular Neuro-Oncology, Department of Neurosurgery, Shaare Zedek-Hebrew University Medical Center, P.O. Box 3235, Jerusalem 9103102, Israel;
| | - Michael Weller
- Laboratory for Molecular Neuro-Oncology, Department of Neurology, University Hospital, University of Zurich, CH-8091 Zurich, Switzerland;
| | - Iris Lavon
- Molecular Neuro-Oncology Laboratory, Leslie and Michael Gaffin Center for Neuro-Oncology, Agnes Ginges Center for Human Neurogenetics, Neurology Department, Hadassah Hebrew University Medical Center, P.O. Box 12000, Jerusalem 91120, Israel; (N.Z.); (M.G.)
| |
Collapse
|
17
|
Nicoll JX, Fry AC, Mosier EM. Androgen and glucocorticoid receptor phosphorylation following resistance exercise and pre-workout supplementation. Steroids 2021; 172:108859. [PMID: 33974920 DOI: 10.1016/j.steroids.2021.108859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/05/2021] [Accepted: 04/24/2021] [Indexed: 11/17/2022]
Abstract
PURPOSE Consumption of caffeine or caffeine containing pre-workout supplements (SUPP) augments steroid hormone responses to resistance exercise (RE). However, the activation of glucocorticoid (GR) and androgen receptors (AR) following RE SUPP has not been investigated. The purpose of this study was to determine the influence of a pre-workout supplement on AR and GR phosphorylation following RE. METHODS In a randomized, counter-balanced, double-blind, placebo-controlled, within-subject crossover study, ten resistance-trained males ((X¯±SD, age = 22 ± 2.4 yrs, hgt = 175 ± 7 cm, body mass = 84.1 ± 11.8 kg) performed four sets of 8 repetitions of barbell back squats at 75% of their 1-repetition maximum (1-RM) with two minutes of rest between sets and a fifth set of barbell back squats at 60% of 1-RM until concentric failure. A SUPP or flavor and color matched placebo (PL) was consumed 60-minutes prior to RE. Vastus lateralis muscle biopsies were obtained prior to supplementation at rest (BL), and ten minutes post-exercise (POST). Biopsies were analyzed for phosphorylated GR (ser134, ser211, and ser226) and phosphorylated AR (ser81, ser213, ser515, ser650) via western blotting. RESULTS pGRser134 decreased, and pGRser226 increased following RE (p < 0.05) with no difference between conditions (p > 0.05). pGRser211 was unchanged after RE (p > 0.05). pARser515 increased, and total AR expression decreased after RE (p < 0.05) in SUPP only. Testosterone and cortisol were not different between SUPP and PL at POST (p > 0.05). CONCLUSION RE influences AR and GR phosphorylation, and SUPP minimally influences this response in the early recovery period.
Collapse
Affiliation(s)
- Justin X Nicoll
- California State University, Northridge, Department of Kinesiology, Northridge, CA, USA.
| | - Andrew C Fry
- University of Kansas, Department of Health, Sport, and Exercise Sciences, Lawrence, KS, USA.
| | - Eric M Mosier
- Northwest Missouri State University, School of Health Science and Wellness, Maryville, MO, USA.
| |
Collapse
|
18
|
|
19
|
Ullah R, Yin Q, Snell AH, Wan L. RAF-MEK-ERK pathway in cancer evolution and treatment. Semin Cancer Biol 2021; 85:123-154. [PMID: 33992782 DOI: 10.1016/j.semcancer.2021.05.010] [Citation(s) in RCA: 235] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/03/2021] [Accepted: 05/06/2021] [Indexed: 12/13/2022]
Abstract
The RAF-MEK-ERK signaling cascade is a well-characterized MAPK pathway involved in cell proliferation and survival. The three-layered MAPK signaling cascade is initiated upon RTK and RAS activation. Three RAF isoforms ARAF, BRAF and CRAF, and their downstream MEK1/2 and ERK1/2 kinases constitute a coherently orchestrated signaling module that directs a range of physiological functions. Genetic alterations in this pathway are among the most prevalent in human cancers, which consist of numerous hot-spot mutations such as BRAFV600E. Oncogenic mutations in this pathway often override otherwise tightly regulated checkpoints to open the door for uncontrolled cell growth and neoplasia. The crosstalk between the RAF-MEK-ERK axis and other signaling pathways further extends the proliferative potential of this pathway in human cancers. In this review, we summarize the molecular architecture and physiological functions of the RAF-MEK-ERK pathway with emphasis on its dysregulations in human cancers, as well as the efforts made to target the RAF-MEK-ERK module using small molecule inhibitors.
Collapse
Affiliation(s)
- Rahim Ullah
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Qing Yin
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Aidan H Snell
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Lixin Wan
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA; Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA.
| |
Collapse
|
20
|
Afshari A, Janfeshan S, Yaghobi R, Roozbeh J, Azarpira N. Covid-19 pathogenesis in prostatic cancer and TMPRSS2-ERG regulatory genetic pathway. INFECTION GENETICS AND EVOLUTION 2020; 88:104669. [PMID: 33301988 PMCID: PMC7720011 DOI: 10.1016/j.meegid.2020.104669] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/09/2020] [Accepted: 12/04/2020] [Indexed: 12/11/2022]
Abstract
Members of Coronaviridae family have been the source of respiratory illnesses. The outbreak of SARS-CoV-2 that produced a severe lung disease in afflicted patients in China and other countries was the reason for the incredible attention paid toward this viral infection. It is known that SARS-CoV-2 is dependent on TMPRSS2 activity for entrance and subsequent infection of the host cells and TMPRSS2 is a host cell molecule that is important for the spread of viruses such as coronaviruses. Different factors can increase the risk of prostate cancer, including older age, a family history of the disease. Androgen receptor (AR) initiates a transcriptional cascade which plays a serious role in both normal and malignant prostate tissues. TMPRSS2 protein is highly expressed in prostate secretory epithelial cells, and its expression is dependent on androgen signals. One of the molecular signs of prostate cancer is TMPRSS2-ERG gene fusion. In TMPRSS2-ERG-positive prostate cancers different patterns of changed gene expression can be detected. The possible molecular relation between fusion positive prostate cancer patients and the increased risk of lethal respiratory viral infections especially SARS-CoV-2 can candidate TMPRSS2 as an attractive drug target. The studies show that some molecules such as nicotinamide, PARP1, ETS and IL-1R can be studied deeper in order to control SARS-CoV-2 infection especially in prostate cancer patients. This review attempts to investigate the possible relation between the gene expression pattern that is produced through TMPRSS2-ERG fusion positive prostate cancer and the possible influence of these fluctuations on the pathogenesis and development of viral infections such as SARS-CoV-2.
Collapse
Affiliation(s)
- Afsoon Afshari
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sahar Janfeshan
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ramin Yaghobi
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jamshid Roozbeh
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Negar Azarpira
- Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
21
|
Gould ML, Nicholson HD. Changes in receptor location affect the ability of oxytocin to stimulate proliferative growth in prostate epithelial cells. Reprod Fertil Dev 2020; 31:1166-1179. [PMID: 31034785 DOI: 10.1071/rd18362] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 02/04/2019] [Indexed: 12/14/2022] Open
Abstract
In normal prostate cells, cell membrane receptors are located within signalling microdomains called caveolae. During cancer progression, caveolae are lost and sequestered receptors move out onto lipid rafts. The aim of this study was to investigate whether a change in the localisation of receptors out of caveolae and onto the cell membrane increased cell proliferation invitro, and to determine whether this is related to changes in the cell signalling pathways. Normal human prostate epithelial cells (PrEC) and androgen-independent (PC3) cancer cells were cultured with 10nM dihydrotestosterone (DHT). The effects of oxytocin (OT) and gonadal steroids on proliferation were assessed using the MTS assay. Androgen receptor (AR) and oxytocin receptor (OTR) expression was identified by immunofluorescence and quantified by western blot. OTR and lipid raft staining was determined using Pearson's correlation coefficient. Protein-protein interactions were detected and the cell signalling pathways identified. Treatment with OT did not affect the proliferation of PrEC. In PC3 cells, OT or androgen alone increased cell proliferation, but together had no effect. In normal cells, OTR localised to the membrane and AR localised to the nucleus, whereas in malignant cells both OTR and AR were identified in the cell membrane. Colocalisation of OTR and AR increased following treatment with androgens. Significantly fewer OTR/AR protein-protein interactions were seen in PrEC. With OT treatment, several cell signalling pathways were activated. Movement of OTR out of caveolae onto lipid rafts is accompanied by activation of alternative signal transduction pathways involved in stimulating increased cell proliferation.
Collapse
Affiliation(s)
- M L Gould
- Anatomy Department, University of Otago, PO Box 913, Dunedin 9054, New Zealand; and Corresponding author.
| | - H D Nicholson
- Anatomy Department, University of Otago, PO Box 913, Dunedin 9054, New Zealand
| |
Collapse
|
22
|
Shorning BY, Dass MS, Smalley MJ, Pearson HB. The PI3K-AKT-mTOR Pathway and Prostate Cancer: At the Crossroads of AR, MAPK, and WNT Signaling. Int J Mol Sci 2020; 21:E4507. [PMID: 32630372 PMCID: PMC7350257 DOI: 10.3390/ijms21124507] [Citation(s) in RCA: 362] [Impact Index Per Article: 72.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/22/2020] [Accepted: 06/22/2020] [Indexed: 12/13/2022] Open
Abstract
Oncogenic activation of the phosphatidylinositol-3-kinase (PI3K), protein kinase B (PKB/AKT), and mammalian target of rapamycin (mTOR) pathway is a frequent event in prostate cancer that facilitates tumor formation, disease progression and therapeutic resistance. Recent discoveries indicate that the complex crosstalk between the PI3K-AKT-mTOR pathway and multiple interacting cell signaling cascades can further promote prostate cancer progression and influence the sensitivity of prostate cancer cells to PI3K-AKT-mTOR-targeted therapies being explored in the clinic, as well as standard treatment approaches such as androgen-deprivation therapy (ADT). However, the full extent of the PI3K-AKT-mTOR signaling network during prostate tumorigenesis, invasive progression and disease recurrence remains to be determined. In this review, we outline the emerging diversity of the genetic alterations that lead to activated PI3K-AKT-mTOR signaling in prostate cancer, and discuss new mechanistic insights into the interplay between the PI3K-AKT-mTOR pathway and several key interacting oncogenic signaling cascades that can cooperate to facilitate prostate cancer growth and drug-resistance, specifically the androgen receptor (AR), mitogen-activated protein kinase (MAPK), and WNT signaling cascades. Ultimately, deepening our understanding of the broader PI3K-AKT-mTOR signaling network is crucial to aid patient stratification for PI3K-AKT-mTOR pathway-directed therapies, and to discover new therapeutic approaches for prostate cancer that improve patient outcome.
Collapse
Affiliation(s)
| | | | | | - Helen B. Pearson
- The European Cancer Stem Cell Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, Wales, UK; (B.Y.S.); (M.S.D.); (M.J.S.)
| |
Collapse
|
23
|
Wenjing H, Shao Y, Yu Y, Huang W, Feng G, Li J. Exendin-4 enhances the sensitivity of prostate cancer to enzalutamide by targeting Akt activation. Prostate 2020; 80:367-375. [PMID: 31967357 DOI: 10.1002/pros.23951] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 12/30/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND Glucagon-like peptide 1 (GLP-1) and its analogs are first-line choices for the treatment of type 2 diabetes mellitus. Recent studies have shown that they exhibit antitumor properties in some tumors. We previously found that a GLP-1 analog, exendin-4 (Ex-4), inhibited the growth of prostate cancer cells through suppressing the PI3K/Akt/mTOR pathway, which is activated in response to enzalutamide treatment and reported to be closely related to resistance to enzalutamide. So we speculated that exendin-4 may enhance the sensitivity of prostate cancer to enzalutamide through inhibiting Akt activation. METHODS LNCap and CWR22RV1 cell lines, as well as mice bearing xenografts formed from the two cells, were used. RESULTS Exendin-4 in combination with enzalutamide dramatically suppressed tumor growth of prostate cancer cells compared to enzalutamide alone; exendin-4 is capable of antagonizing enzalutamide-induced invasion and migration of both prostate cancer cells (P < .05). Furthermore, the combination treatment significantly reduced Akt and mTOR levels that were triggered by enzalutamide administration, caused a further decrease in nuclear AR localization compared with the enzalutamide as a monotherapy (P < .5), though exendin-4 treatment alone showed no effect on nuclear AR. CONCLUSION Our study demonstrated that exendin-4 alleviated resistance to enzalutamide, and suggested that exendin-4 combined with enzalutamide may be a more efficacious treatment for patients with advanced prostate cancer.
Collapse
Affiliation(s)
- He Wenjing
- Institute of Urology, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yuanyuan Shao
- Department of Oncology, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yi Yu
- Department of Oncology, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wei Huang
- Department of Oncology, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Guoliang Feng
- Department of Oncology, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Junhe Li
- Department of Oncology, the First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
24
|
You L, Lin Q, Zhao J, Shi F, Young KH, Qian W. Whole-exome sequencing identifies novel somatic alterations associated with outcomes in idiopathic multicentric Castleman disease. Br J Haematol 2020; 188:e64-e67. [PMID: 31863597 DOI: 10.1111/bjh.16330] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Liangshun You
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
- Malignant Lymphoma Diagnosis and Therapy Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qingqing Lin
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Malignant Lymphoma Diagnosis and Therapy Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jing Zhao
- Department of Pathology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Fangjing Shi
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ken H Young
- Department of Pathology and Hematopathology Division, Duke University School of Medicine, Duke Medical Center and Cancer Institute, Durham, NC, USA
| | - Wenbin Qian
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
- Malignant Lymphoma Diagnosis and Therapy Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
25
|
Rossini A, Giussani M, Ripamonti F, Aiello P, Regondi V, Balsari A, Triulzi T, Tagliabue E. Combined targeting of EGFR and HER2 against prostate cancer stem cells. Cancer Biol Ther 2020; 21:463-475. [PMID: 32089070 DOI: 10.1080/15384047.2020.1727702] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Progression of prostate cancer has been associated with EGFR and HER2 activation and to tumor-initiating cells contribution toward chemotherapy resistance. We investigated the efficacy of a dual intervention against EGFR and HER2 to deplete the tumor-initiating cells, optimize the chemotherapy management and prevent the progression of castration-resistant prostate cancer (CRPC) cells. Using DU145, PC3, and 22Rv1 CRPC cell lines, biochemical analysis revealed activation of EGFR, HER2, MAPK, and STAT3 in DU145 and 22Rv1, and AKT and SRC in DU145 and PC-3. pSTAT3 nuclear staining was observed in DU145 xenografts and in 12 out of 14 CRPC specimens. The in vivo dual targeting of ErbB receptors with Cetuximab and Trastuzumab combined with chemotherapy caused an effective antitumor response in DU145 xenografted mice displaying STAT3 activation; conversely PC-3 bearing mice experienced tumor relapse. The potentiating of in vivo cytotoxic effect in DU145 model was accompanied by a significant decrease of prostatosphere-forming capacity assessed in vitro on residual tumor cells. Additionally, combined treatment in vitro with Cetuximab, Trastuzumab and chemotherapy negatively affected DU145 and 22Rv1 sphere formation, suggesting the critical function of ErbB receptors for tumor-initiating cells proliferation; no effect on PC-3 clonogenic potential was observed, indicating that other receptors than EGFR and HER2 may sustain PC3 tumor-initiating cells. These findings provided the preclinical evidence that the dual inhibition of EGFR and HER2 by targeting tumor-initiating cells may improve the efficacy of the current chemotherapy regimen, bringing benefits especially to castration-resistant patients with activated STAT3, and preventing disease progression.
Collapse
Affiliation(s)
- Anna Rossini
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Marta Giussani
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Francesca Ripamonti
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Piera Aiello
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Viola Regondi
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Andrea Balsari
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy.,Dipartimento Di Scienze Biomediche per La Salute, Università Degli Studi Di Milano, Milan, Italy
| | - Tiziana Triulzi
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Elda Tagliabue
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| |
Collapse
|
26
|
Varnosfaderani ZG, Emamzadeh R, Nazari M, Zarean M. Detection of a prostate cancer cell line using a bioluminescent affiprobe: An attempt to develop a new molecular probe for ex vivo studies. Int J Biol Macromol 2019; 138:755-763. [DOI: 10.1016/j.ijbiomac.2019.07.085] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 06/26/2019] [Accepted: 07/11/2019] [Indexed: 11/16/2022]
|
27
|
Nicoll JX, Fry AC, Mosier EM, Olsen LA, Sontag SA. MAPK, androgen, and glucocorticoid receptor phosphorylation following high-frequency resistance exercise non-functional overreaching. Eur J Appl Physiol 2019; 119:2237-2253. [DOI: 10.1007/s00421-019-04200-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 07/29/2019] [Indexed: 12/26/2022]
|
28
|
Gesmundo I, Di Blasio L, Banfi D, Villanova T, Fanciulli A, Favaro E, Gamba G, Musuraca C, Rapa I, Volante M, Munegato S, Papotti M, Gontero P, Primo L, Ghigo E, Granata R. Proton pump inhibitors promote the growth of androgen-sensitive prostate cancer cells through ErbB2, ERK1/2, PI3K/Akt, GSK-3β signaling and inhibition of cellular prostatic acid phosphatase. Cancer Lett 2019; 449:252-262. [PMID: 30790678 DOI: 10.1016/j.canlet.2019.02.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 02/08/2019] [Accepted: 02/14/2019] [Indexed: 12/13/2022]
Abstract
Prostate cancer (PCa) is one of the most common cancer in men. Although hormone-sensitive PCa responds to androgen-deprivation, there are no effective therapies for castration-resistant PCa. It has been recently suggested that proton pump inhibitors (PPIs) may increase the risk of certain cancers; however, association with PCa remains elusive. Here, we evaluated the tumorigenic activities of PPIs in vitro, in PCa cell lines and epithelial cells from benign prostatic hyperplasia (BPH) and in vivo, in PCa mice xenografts. PPIs increased survival and proliferation, and inhibited apoptosis in LNCaP cells. These effects were attenuated or absent in androgen-insensitive DU-145 and PC3 cells, respectively. Specifically, omeprazole (OME) promoted cell cycle progression, increased c-Myc expression, ErbB2 activity and PSA secretion. Furthermore, OME induced the phosphorylation of MAPK-ERK1/2, PI3K/Akt and GSK-3β, and blunted the expression and activity of cellular prostatic acid phosphatase. OME also increased survival, proliferation and PSA levels in BPH cells. In vivo, OME promoted tumor growth in mice bearing LNCaP xenografts. Our results indicate that PPIs display tumorigenic activities in PCa cells, suggesting that their long-term administration in patients should be carefully monitored.
Collapse
Affiliation(s)
- Iacopo Gesmundo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin and Città Della Salute e Della Scienza Hospital, Turin, 10126, Italy
| | - Laura Di Blasio
- Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, 10060, Italy; Department of Oncology, University of Turin, Turin, Italy
| | - Dana Banfi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin and Città Della Salute e Della Scienza Hospital, Turin, 10126, Italy
| | - Tania Villanova
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin and Città Della Salute e Della Scienza Hospital, Turin, 10126, Italy
| | - Alessandro Fanciulli
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin and Città Della Salute e Della Scienza Hospital, Turin, 10126, Italy
| | - Enrica Favaro
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin and Città Della Salute e Della Scienza Hospital, Turin, 10126, Italy
| | - Giacomo Gamba
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin and Città Della Salute e Della Scienza Hospital, Turin, 10126, Italy
| | - Chiara Musuraca
- Department of Oncology, University of Turin, Turin, Italy; Città Della Salute e Della Scienza Hospital, Turin, 10126, Italy
| | - Ida Rapa
- Department of Oncology, University of Turin, San Luigi Hospital, Orbassano, Turin, 10043, Italy
| | - Marco Volante
- Department of Oncology, University of Turin, San Luigi Hospital, Orbassano, Turin, 10043, Italy
| | - Stefania Munegato
- Division of Urology, Department of Surgical Sciences, University of Turin and Città Della Salute e Della Scienza Hospital, Turin, 10126, Italy
| | - Mauro Papotti
- Department of Oncology, University of Turin, Turin, Italy; Città Della Salute e Della Scienza Hospital, Turin, 10126, Italy
| | - Paolo Gontero
- Division of Urology, Department of Surgical Sciences, University of Turin and Città Della Salute e Della Scienza Hospital, Turin, 10126, Italy
| | - Luca Primo
- Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, 10060, Italy; Department of Oncology, University of Turin, Turin, Italy
| | - Ezio Ghigo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin and Città Della Salute e Della Scienza Hospital, Turin, 10126, Italy
| | - Riccarda Granata
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin and Città Della Salute e Della Scienza Hospital, Turin, 10126, Italy.
| |
Collapse
|
29
|
Bousset L, Rambur A, Fouache A, Bunay J, Morel L, Lobaccaro JMA, Baron S, Trousson A, de Joussineau C. New Insights in Prostate Cancer Development and Tumor Therapy: Modulation of Nuclear Receptors and the Specific Role of Liver X Receptors. Int J Mol Sci 2018; 19:E2545. [PMID: 30154328 PMCID: PMC6164771 DOI: 10.3390/ijms19092545] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/08/2018] [Accepted: 08/09/2018] [Indexed: 12/14/2022] Open
Abstract
Prostate cancer (PCa) incidence has been dramatically increasing these last years in westernized countries. Though localized PCa is usually treated by radical prostatectomy, androgen deprivation therapy is preferred in locally advanced disease in combination with chemotherapy. Unfortunately, PCa goes into a castration-resistant state in the vast majority of the cases, leading to questions about the molecular mechanisms involving the steroids and their respective nuclear receptors in this relapse. Interestingly, liver X receptors (LXRα/NR1H3 and LXRβ/NR1H2) have emerged as new actors in prostate physiology, beyond their historical roles of cholesterol sensors. More importantly LXRs have been proposed to be good pharmacological targets in PCa. This rational has been based on numerous experiments performed in PCa cell lines and genetic animal models pointing out that using selective liver X receptor modulators (SLiMs) could actually be a good complementary therapy in patients with a castration resistant PCa. Hence, this review is focused on the interaction among the androgen receptors (AR/NR3C4), estrogen receptors (ERα/NR3A1 and ERβ/NR3A2), and LXRs in prostate homeostasis and their putative pharmacological modulations in parallel to the patients' support.
Collapse
Affiliation(s)
- Laura Bousset
- Université Clermont Auvergne, GReD, CNRS UMR 6293, INSERM U1103, 28, place Henri Dunant, BP38, F63001 Clermont-Ferrand, France.
- Centre de Recherche en Nutrition Humaine d'Auvergne, 58 Boulevard Montalembert, F-63009 Clermont-Ferrand, France.
| | - Amandine Rambur
- Université Clermont Auvergne, GReD, CNRS UMR 6293, INSERM U1103, 28, place Henri Dunant, BP38, F63001 Clermont-Ferrand, France.
- Centre de Recherche en Nutrition Humaine d'Auvergne, 58 Boulevard Montalembert, F-63009 Clermont-Ferrand, France.
| | - Allan Fouache
- Université Clermont Auvergne, GReD, CNRS UMR 6293, INSERM U1103, 28, place Henri Dunant, BP38, F63001 Clermont-Ferrand, France.
- Centre de Recherche en Nutrition Humaine d'Auvergne, 58 Boulevard Montalembert, F-63009 Clermont-Ferrand, France.
| | - Julio Bunay
- Université Clermont Auvergne, GReD, CNRS UMR 6293, INSERM U1103, 28, place Henri Dunant, BP38, F63001 Clermont-Ferrand, France.
- Centre de Recherche en Nutrition Humaine d'Auvergne, 58 Boulevard Montalembert, F-63009 Clermont-Ferrand, France.
| | - Laurent Morel
- Université Clermont Auvergne, GReD, CNRS UMR 6293, INSERM U1103, 28, place Henri Dunant, BP38, F63001 Clermont-Ferrand, France.
- Centre de Recherche en Nutrition Humaine d'Auvergne, 58 Boulevard Montalembert, F-63009 Clermont-Ferrand, France.
| | - Jean-Marc A Lobaccaro
- Université Clermont Auvergne, GReD, CNRS UMR 6293, INSERM U1103, 28, place Henri Dunant, BP38, F63001 Clermont-Ferrand, France.
- Centre de Recherche en Nutrition Humaine d'Auvergne, 58 Boulevard Montalembert, F-63009 Clermont-Ferrand, France.
| | - Silvère Baron
- Université Clermont Auvergne, GReD, CNRS UMR 6293, INSERM U1103, 28, place Henri Dunant, BP38, F63001 Clermont-Ferrand, France.
- Centre de Recherche en Nutrition Humaine d'Auvergne, 58 Boulevard Montalembert, F-63009 Clermont-Ferrand, France.
| | - Amalia Trousson
- Université Clermont Auvergne, GReD, CNRS UMR 6293, INSERM U1103, 28, place Henri Dunant, BP38, F63001 Clermont-Ferrand, France.
- Centre de Recherche en Nutrition Humaine d'Auvergne, 58 Boulevard Montalembert, F-63009 Clermont-Ferrand, France.
| | - Cyrille de Joussineau
- Université Clermont Auvergne, GReD, CNRS UMR 6293, INSERM U1103, 28, place Henri Dunant, BP38, F63001 Clermont-Ferrand, France.
- Centre de Recherche en Nutrition Humaine d'Auvergne, 58 Boulevard Montalembert, F-63009 Clermont-Ferrand, France.
| |
Collapse
|
30
|
Yeoh CC, Dabab N, Rigby E, Chhikara R, Akaev I, Gomez RS, Fonseca F, Brennan PA, Rahimi S. Androgen receptor in salivary gland carcinoma: A review of an old marker as a possible new target. J Oral Pathol Med 2018; 47:691-695. [DOI: 10.1111/jop.12741] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Chit Cheng Yeoh
- Department of Oncology; Queen Alexandra Hospital; Portsmouth UK
| | - Nedal Dabab
- Department of Oncology; Queen Alexandra Hospital; Portsmouth UK
| | - Elyse Rigby
- Department of Oncology; Queen Alexandra Hospital; Portsmouth UK
| | - Ritu Chhikara
- Pathology Centre-Histopathology; Queen Alexandra Hospital; Portsmouth UK
| | - Iolia Akaev
- School of Pharmacy and Biomedical Science; University of Portsmouth; Portsmouth UK
| | - Ricardo Santiago Gomez
- Department of Oral Surgery and Pathology; School of Dentistry; Universidade Federal de Minas Gerais; Belo Horizonte Brazil
| | - Felipe Fonseca
- Department of Oral Surgery and Pathology; School of Dentistry; Universidade Federal de Minas Gerais; Belo Horizonte Brazil
| | - Peter A. Brennan
- Department of Oral and Maxillofacial Surgery; Queen Alexandra Hospital; Portsmouth UK
| | - Siavash Rahimi
- Pathology Centre-Histopathology; Queen Alexandra Hospital; Portsmouth UK
- School of Pharmacy and Biomedical Science; University of Portsmouth; Portsmouth UK
| |
Collapse
|
31
|
Mandel A, Larsson P, Sarwar M, Semenas J, Syed Khaja AS, Persson JL. The interplay between AR, EGF receptor and MMP-9 signaling pathways in invasive prostate cancer. Mol Med 2018; 24:34. [PMID: 30134822 PMCID: PMC6020326 DOI: 10.1186/s10020-018-0035-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 06/11/2018] [Indexed: 12/17/2022] Open
Abstract
Background Metastatic Prostate cancer (PCa) cells have gained survival and invasive advantages. Epidermal growth factor (EGF) receptor is a receptor tyrosine kinase, which may mediate signalling to promote progression and invasion of various cancers. In this study, we uncovered the molecular mechanisms underlying the interconnection among the androgen receptor (AR), matrix metalloproteinase-9 (MMP9) and EGFR in promoting PCa progression. Methods Immunohistochemical analysis of the tissue microarrays consisting of primary and metastatic PCa tissues was performed. The clinical importance of EGFR and its association with survivals were analyzed using three cohorts from MSKCC Prostate Oncogenome Project dataset (For primary tumors, n = 181; for metastatic tumors n = 37) and The Cancer Genome Atlas Prostate Adenocarcinoma Provisional dataset (n = 495). Targeted overexpression or inhibition of the proteins of interests was introduced into PCa cell lines. Treatment of PCa cell lines with the compounds was conducted. Immunoblot analysis was performed. Results We showed that AR, MMP-9 and EGFR are interconnect factors, which may cooperatively promote PCa progression. Altered EGFR expression was associated with poor disease-free survival in PCa patients. Induced overexpression of AR led to an increase in the expression of EGFR, p-GSK-3β and decrease in p27 expression in PCa cell lines in the presence of androgen stimulation. Overexpression of MMP9 significantly induced EGFR expression in PCa cells. Inhibition of PIP5K1α, a lipid kinase that acts upstream of PI3K/AKT greatly reduced expressions of AR, MMP-9 and EGFR. Conclusions Our findings also suggest that PCa cells may utilize AR, EGFR and MMP-9 pathways in androgen-dependent as well as in castration-resistant conditions. Our data suggest a new therapeutic potential to block cancer metastasis by targeting AR, EGFR and MMP-9 pathways in subsets of PCa patients.
Collapse
Affiliation(s)
- Anna Mandel
- Department of Molecular Biology, Umeå University, 901 87, Umeå, Sweden
| | - Per Larsson
- Department of Molecular Biology, Umeå University, 901 87, Umeå, Sweden
| | - Martuza Sarwar
- Division of Experimental Cancer Research, Department of Translational Medicine, Clinical Research Centre, Lund University, Jan Waldenströms gatan 35, 205 02, Malmö, Sweden
| | - Julius Semenas
- Department of Molecular Biology, Umeå University, 901 87, Umeå, Sweden
| | | | - Jenny L Persson
- Department of Molecular Biology, Umeå University, 901 87, Umeå, Sweden. .,Division of Experimental Cancer Research, Department of Translational Medicine, Clinical Research Centre, Lund University, Jan Waldenströms gatan 35, 205 02, Malmö, Sweden.
| |
Collapse
|
32
|
Di Lorenzo G, Autorino R, De Laurentiis M, Cindolo L, D'Armiento M, Bianco AR, De Placido S. Her-2/Neu Receptor in Prostate Cancer Development and Progression to Androgen Independence. TUMORI JOURNAL 2018; 90:163-70. [PMID: 15237576 DOI: 10.1177/030089160409000201] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Development of prostate cancer and progression to androgen-independent disease is correlated with increased expression of growth factors and receptors capable of establishing autocrine and/or paracrine growth-stimulatory loops. A thorough review was made of the current literature and recent abstract presentations at scientific meetings focusing on the role of the HER-2/neu (c-erbB2) receptor in prostate cancer and the potential clinical usefulness of its specific inhibitors. In the past 10 years, conflicting results on HER-2/neu expression in prostate cancer have been reported. More recently, four studies have shown experimental evidence of HER-2/neu in the development of prostate cancer and, more specifically, in the progression to a hormone-refractory clinical behavior. Furthermore, it has been proposed that HER-2 family and androgen receptors function synergistically in the absence of androgen, which suggests a crosstalk between the HER-2/neu and androgen receptor pathways. Finally, clinical trials are in progress in prostate cancer patients to test novel agents that selectively interfere with HER-2/neu activity.
Collapse
Affiliation(s)
- Giuseppe Di Lorenzo
- Dipartimento di Endocrinologia e Oncologia Molecolare e Clinica, Università degli Studi Federico II, Naples, Italy.
| | | | | | | | | | | | | |
Collapse
|
33
|
Lai HC, Yeh CC, Jeng LB, Huang SF, Liao PY, Lei FJ, Cheng WC, Hsu CL, Cai X, Chang C, Ma WL. Androgen receptor mitigates postoperative disease progression of hepatocellular carcinoma by suppressing CD90+ populations and cell migration and by promoting anoikis in circulating tumor cells. Oncotarget 2018; 7:46448-46465. [PMID: 27340775 PMCID: PMC5216809 DOI: 10.18632/oncotarget.10186] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 05/28/2016] [Indexed: 02/06/2023] Open
Abstract
Purpose Although hepatectomy and liver transplantation surgery for hepatocellular carcinoma (HCC) are effective treatment modalities, the risk of recurrence remains high, particularly in patients with a high number of circulating tumor cells (CTCs) expressing cancer stem/progenitor cell markers. Androgen receptor (AR) signaling has been shown to suppress HCC metastasis in rodent models of HCC. In this study, we investigated whether AR is associated with postoperative HCC recurrence. Experimental Design CTCs were obtained from patients with HCC who had undergone hepatectomy to investigate whether they are associated with disease outcome. AR knockout was introduced in two mouse models of spontaneous HCC (carcinogen- and hepatitis B virus-related HCC) to delineate the role that AR plays in HCC recurrence. Biological systems analysis was used to investigate the cellular and molecular mechanisms. Results We found that the expression of AR in CTCs was negatively associated with HCC recurrence/progression after hepatectomy. Our results suggest that AR-mediated suppression of HCC recurrence/progression is governed by a three-pronged mechanism. First, AR suppresses the expression of CD90 in CTCs by upregulating Histone 3H2A. Second, AR suppresses cell migration at the transcriptome level. Third, AR promotes anoikis of CTCs via dysregulation of cytoskeletal adsorption. Conclusions The results indicate that AR expression may be the gatekeeper of postoperative HCC recurrence. Therefore, targeting AR in presurgical down-staging procedures may serve as a secondary prevention measure against HCC recurrence in the future.
Collapse
Affiliation(s)
- Hsueh-Chou Lai
- Graduate Institution of Clinical Medical Science, and Graduate Institution of Cancer Biology, China Medical University, Taichung 40403, Taiwan.,Sex Hormone Research Center, Organ Transplantation Center, Research Center for Tumor Medical Science, and Department of Gastroenterology, China Medical University/Hospital, Taichung 40403, Taiwan
| | - Chun-Chieh Yeh
- Graduate Institution of Clinical Medical Science, and Graduate Institution of Cancer Biology, China Medical University, Taichung 40403, Taiwan.,Sex Hormone Research Center, Organ Transplantation Center, Research Center for Tumor Medical Science, and Department of Gastroenterology, China Medical University/Hospital, Taichung 40403, Taiwan
| | - Long-Bin Jeng
- Graduate Institution of Clinical Medical Science, and Graduate Institution of Cancer Biology, China Medical University, Taichung 40403, Taiwan
| | - Shang-Fen Huang
- Sex Hormone Research Center, Organ Transplantation Center, Research Center for Tumor Medical Science, and Department of Gastroenterology, China Medical University/Hospital, Taichung 40403, Taiwan
| | - Pei-Ying Liao
- Sex Hormone Research Center, Organ Transplantation Center, Research Center for Tumor Medical Science, and Department of Gastroenterology, China Medical University/Hospital, Taichung 40403, Taiwan
| | - Fu-Ju Lei
- Graduate Institution of Clinical Medical Science, and Graduate Institution of Cancer Biology, China Medical University, Taichung 40403, Taiwan.,Sex Hormone Research Center, Organ Transplantation Center, Research Center for Tumor Medical Science, and Department of Gastroenterology, China Medical University/Hospital, Taichung 40403, Taiwan
| | - Wei-Chun Cheng
- Graduate Institution of Clinical Medical Science, and Graduate Institution of Cancer Biology, China Medical University, Taichung 40403, Taiwan.,Sex Hormone Research Center, Organ Transplantation Center, Research Center for Tumor Medical Science, and Department of Gastroenterology, China Medical University/Hospital, Taichung 40403, Taiwan
| | - Cheng-Lung Hsu
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung University/Memorial Hospital, Taoyuan 333, Taiwan
| | - Xiujun Cai
- Chawnshang Chang Liver Cancer Center, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou 310016, China
| | - Chawnshang Chang
- Sex Hormone Research Center, Organ Transplantation Center, Research Center for Tumor Medical Science, and Department of Gastroenterology, China Medical University/Hospital, Taichung 40403, Taiwan.,Chawnshang Chang Liver Cancer Center, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou 310016, China.,George Whipple Laboratory for Cancer Research, Department of Pathology, The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY 14623, USA
| | - Wen-Lung Ma
- Graduate Institution of Clinical Medical Science, and Graduate Institution of Cancer Biology, China Medical University, Taichung 40403, Taiwan.,Sex Hormone Research Center, Organ Transplantation Center, Research Center for Tumor Medical Science, and Department of Gastroenterology, China Medical University/Hospital, Taichung 40403, Taiwan
| |
Collapse
|
34
|
Di Vizio D, Solomon KR, Freeman MR. Cholesterol and Cholesterol-Rich Membranes in Prostate Cancer: An Update. TUMORI JOURNAL 2018; 94:633-9. [DOI: 10.1177/030089160809400501] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cells maintain normal structure and function by responding appropriately to cues from the surrounding milieu. Extracellular stimuli are transduced from the surface through the plasma membrane by a complex series of interactions between ligands, their receptors and intracellular signaling partners (e.g., kinases, G proteins). Cholesterol-enriched membrane microdomains, generally referred to as “lipid rafts”, exist within the lipid bilayer of all mammalian cells and play an important role in signaling from the cell surface to various subcellular compartments. Lipid rafts have also been implicated in tumor growth and aggressiveness. Epidemiological evidence suggests that the modern Western diet, which contains substantial levels of cholesterol and other fatty substances, promotes prostate cancer progression. Consistent with this idea, prolonged inhibition of the cholesterol synthesis pathway by pharmacologic intervention in men has recently been associated with reduction in risk of advanced prostate cancer. In this review, we discuss the possibility that membrane cholesterol promotes prostate cancer progression by a mechanism that involves dysregulation of lipid raft-resident signaling complexes. This hypothesis provides new avenues for mechanistic studies as well as therapeutic intervention.
Collapse
Affiliation(s)
- Dolores Di Vizio
- The Urological Diseases Research Center, Department of Urology, Children's Hospital Boston, Harvard Medical School, Boston, MA (USA)
- Department of Surgery, Harvard Medical School, Boston, MA (USA)
| | - Keith R Solomon
- The Urological Diseases Research Center, Department of Urology, Children's Hospital Boston, Harvard Medical School, Boston, MA (USA)
- The Urological Diseases Research Center, Department of Orthopaedic Surgery, Children's Hospital Boston, Harvard Medical School, Boston, MA (USA)
- Department of Orthopaedic Surgery, Harvard Medical School, Boston, MA (USA)
| | - Michael R Freeman
- The Urological Diseases Research Center, Department of Urology, Children's Hospital Boston, Harvard Medical School, Boston, MA (USA)
- Department of Surgery, Harvard Medical School, Boston, MA (USA)
| |
Collapse
|
35
|
Neto AS, Tobias-Machado M, Wroclawski ML, Fonseca FLA, Pompeo ACL, Del Giglio A. Molecular Oncogenesis of Prostate Adenocarcinoma: Role of the Human Epidermal Growth Factor Receptor 2 (HER-2/neu). TUMORI JOURNAL 2018; 96:645-9. [DOI: 10.1177/030089161009600501] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The potential mechanisms involving the genesis and growth of androgen-independent prostate cancer include super-expression of the androgen receptor (AR), in an attempt to compensate for the low androgenic plasma levels and mutations of this specific receptor, which could determine resistance to anti-androgenic therapy. However, most advanced prostate tumors have no mutations or amplifications of the AR, suggesting a potential role of non-androgenic growth factors, including epidermal growth factor (EGF), transforming growth factor α, insulin-like growth factor (IGF-1) and fibroblast growth factor. More specifically, these factors, and their receptors like EGFR (HER-1) and HER-2/neu, through paracrine and autocrine mechanisms, may contribute to the proliferation and growth of prostate cancer. Free full text available at www.tumorionline.it
Collapse
Affiliation(s)
- Ary Serpa Neto
- Urologic Oncology Division, Department of Urology, ABC Medical School (FMABC), Santo André
| | - Marcos Tobias-Machado
- Urologic Oncology Division, Department of Urology, ABC Medical School (FMABC), Santo André
- Research Institute, Albert Einstein Jewish Hospital (IEP-HIAE), São Paulo
| | - Marcelo Langer Wroclawski
- Urologic Oncology Division, Department of Urology, ABC Medical School (FMABC), Santo André
- Research Institute, Albert Einstein Jewish Hospital (IEP-HIAE), São Paulo
| | - Fernando Luiz Affonso Fonseca
- Research Institute, Albert Einstein Jewish Hospital (IEP-HIAE), São Paulo
- Oncology Division, Department of Clinical Oncology and Hematology, ABC Medicine School (FMABC), Santo André, Brazil
| | | | - Auro Del Giglio
- Research Institute, Albert Einstein Jewish Hospital (IEP-HIAE), São Paulo
- Oncology Division, Department of Clinical Oncology and Hematology, ABC Medicine School (FMABC), Santo André, Brazil
| |
Collapse
|
36
|
Xu D, Chen Q, Liu Y, Wen X. Baicalein suppresses the androgen receptor (AR)-mediated prostate cancer progression via inhibiting the AR N-C dimerization and AR-coactivators interaction. Oncotarget 2017; 8:105561-105573. [PMID: 29285272 PMCID: PMC5739659 DOI: 10.18632/oncotarget.22319] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 06/28/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Androgen receptor (AR) plays a critical role in prostate cancer (PCa) development and progression. Androgen deprivation therapy with antiandrogens to reduce androgen biosynthesis or prevent androgens from binding to AR are widely used to suppress AR-mediated PCa growth. However, most of ADT may eventually fail with development of the castration resistance after 12-24 months. Here we found that a natural product baicalein can effectively suppress the PCa progression via targeting the androgen-induced AR transactivation with little effect to AR protein expression. METHODS PCa cells including LNCaP, CWR22Rv1, C4-2, PC-3, and DU145, were treated with baicalein and luciferase assay was used to evaluate their effect on the AR transactivation. Cell growth and IC50 were determined by MTT assay after 48 hrs treatment. RT-PCR was used to evaluate the mRNA levels of AR target genes including PSA, TMPRSS2, and TMEPA1. Western blot was used to determine AR and PSA protein expression. RESULTS The natural product of baicalein can selectively inhibit AR transactivation with little effect on the other nuclear receptors, including ERα, and GR. At a low concentration, 2.5 μM of baicalein effectively suppresses the growth of AR-positive PCa cells, and has little effect on AR-negative PCa cells. Mechanism dissection suggest that baicalein can suppress AR target genes (PSA, TMPRSS2, and TMEPA1) expression in both androgen responsive LNCaP cells and castration resistant CWR22Rv1 cells, that may involve the inhibiting the AR N/C dimerization and AR-coactivators interaction. CONCLUSIONS Baicalein may be developed as an effective anti-AR therapy via its ability to inhibit AR transactivation and AR-mediated PCa cell growth.
Collapse
Affiliation(s)
- Defeng Xu
- School of Pharmaceutical Engineering and Life Sciences, Changzhou University, Changzhou, Jiangsu 213164, P.R. China
| | - Qiulu Chen
- School of Pharmaceutical Engineering and Life Sciences, Changzhou University, Changzhou, Jiangsu 213164, P.R. China
| | - Yalin Liu
- School of Pharmaceutical Engineering and Life Sciences, Changzhou University, Changzhou, Jiangsu 213164, P.R. China
| | - Xingqiao Wen
- Department of Urology, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong 518100, P.R. China
| |
Collapse
|
37
|
Targeting androgen receptor versus targeting androgens to suppress castration resistant prostate cancer. Cancer Lett 2017; 397:133-143. [DOI: 10.1016/j.canlet.2017.03.022] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 03/11/2017] [Accepted: 03/13/2017] [Indexed: 12/31/2022]
|
38
|
Momtazi-borojeni AA, Abdollahi E, Ghasemi F, Caraglia M, Sahebkar A. The novel role of pyrvinium in cancer therapy. J Cell Physiol 2017; 233:2871-2881. [DOI: 10.1002/jcp.26006] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 05/11/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Amir A. Momtazi-borojeni
- Nanotechnology Research Center; Bu-Ali Research Institute; Mashhad University of Medical Sciences; Mashhad Iran
- Faculty of Medicine; Department of Medical Biotechnology; Student Research Committee; Mashhad University of Medical Sciences; Mashhad Iran
| | - Elham Abdollahi
- Department of Medical Immunology; School of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
- Student Research Committee; Mashhad University of Medical Sciences; Mashhad Iran
| | - Faezeh Ghasemi
- Faculty of Medicine; Department of Medical Biotechnology; Arak University of Medical Sciences; Arak Iran
| | - Michele Caraglia
- Department of Biochemistry; Biophysics and General Pathology; University of Campania “L. Vanvitelli”; Via L. De Crecchio; Naples Italy
| | - Amirhossein Sahebkar
- Biotechnology Research Center; Mashhad University of Medical Sciences; Mashhad Iran
| |
Collapse
|
39
|
Lu S, Dong Z. Overexpression of secretory phospholipase A2-IIa supports cancer stem cell phenotype via HER/ERBB-elicited signaling in lung and prostate cancer cells. Int J Oncol 2017; 50:2113-2122. [PMID: 28440478 DOI: 10.3892/ijo.2017.3964] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 04/10/2017] [Indexed: 11/06/2022] Open
Abstract
Resistance to conventional chemotherapies remains a significant clinical challenge in treatment of cancer. The cancer stem cells (CSCs) have properties necessary for tumor initiation, resistance to therapy, and progression. HER/ERBB‑elicited signaling supports CSC properties. Our previous studies revealed that secretory phospholipase A2 group IIa (sPLA2‑IIa) is overexpressed in both prostate and lung cancer cells, leading to an aberrant high level in the interstitial fluid, i.e., tumor microenvironment and blood. HER/ERBB-PI3K-Akt-NF-κB signaling stimulates sPLA2‑IIa overexpression, and in turn, sPLA2‑IIa activates EGFR family receptors and HER/ERBB-elicited signaling and stimulates sPLA2‑IIa overexpression in a positive feedback manner. The present study determined the molecular mechanisms of sPLA2‑IIa in stimulating HER/ERBB-elicited signaling and supporting CSC properties. We found that sPLA2‑IIa binds both EGFR and HER3 demonstrated by co-immunoprecipitation experiments and also indirectly interacts with HER2, suggesting that sPLA2‑IIa functions as a ligand for both EGFR and HER3. Furthermore, both side population CSCs from non-small cell lung cancer (NSCLC) A549 and H1975 cells and ALDH1‑high CSCs from castration-resistant prostate cancer (CRPC) 22Rv1 cells overexpress sPLA2‑IIa and produce tumors when inoculated into subcutis of nude mice. Given an aberrant high level of sPLA2‑IIa in the tumor microenvironment that should be much higher than that in the blood, our findings support the notion that sPLA2‑IIa functions as a ligand for EGFR family receptors and supports CSC properties via HER/ERBB-elicited signaling, which may contribute to resistance to therapy and cancer progression.
Collapse
Affiliation(s)
- Shan Lu
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Zhongyun Dong
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
40
|
Ramalingam S, Ramamurthy VP, Njar VCO. Dissecting major signaling pathways in prostate cancer development and progression: Mechanisms and novel therapeutic targets. J Steroid Biochem Mol Biol 2017; 166:16-27. [PMID: 27481707 PMCID: PMC7371258 DOI: 10.1016/j.jsbmb.2016.07.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 07/08/2016] [Accepted: 07/12/2016] [Indexed: 12/19/2022]
Abstract
Prostate cancer (PCa) is the most frequently diagnosed non-cutaneous malignancy and leading cause of cancer mortality in men. At the initial stages, prostate cancer is dependent upon androgens for their growth and hence effectively combated by androgen deprivation therapy (ADT). However, most patients eventually recur with an androgen deprivation-resistant phenotype, referred to as castration-resistant prostate cancer (CRPC), a more aggressive form for which there is no effective therapy presently available. The current review is an attempt to cover and establish an understanding of some major signaling pathways implicated in prostate cancer development and castration-resistance, besides addressing therapeutic strategies that targets the key signaling mechanisms.
Collapse
Affiliation(s)
- Senthilmurugan Ramalingam
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA; Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA
| | - Vidya P Ramamurthy
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA; Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA
| | - Vincent C O Njar
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA; Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA.
| |
Collapse
|
41
|
AR-Signaling in Human Malignancies: Prostate Cancer and Beyond. Cancers (Basel) 2017; 9:cancers9010007. [PMID: 28085048 PMCID: PMC5295778 DOI: 10.3390/cancers9010007] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/04/2017] [Accepted: 01/05/2017] [Indexed: 12/11/2022] Open
Abstract
In the 1940s Charles Huggins reported remarkable palliative benefits following surgical castration in men with advanced prostate cancer, and since then the androgen receptor (AR) has remained the main therapeutic target in this disease. Over the past couple of decades, our understanding of AR-signaling biology has dramatically improved, and it has become apparent that the AR can modulate a number of other well-described oncogenic signaling pathways. Not surprisingly, mounting preclinical and epidemiologic data now supports a role for AR-signaling in promoting the growth and progression of several cancers other than prostate, and early phase clinical trials have documented preliminary signs of efficacy when AR-signaling inhibitors are used in several of these malignancies. In this article, we provide an overview of the evidence supporting the use of AR-directed therapies in prostate as well as other cancers, with an emphasis on the rationale for targeting AR-signaling across tumor types.
Collapse
|
42
|
Hoang DT, Iczkowski KA, Kilari D, See W, Nevalainen MT. Androgen receptor-dependent and -independent mechanisms driving prostate cancer progression: Opportunities for therapeutic targeting from multiple angles. Oncotarget 2017; 8:3724-3745. [PMID: 27741508 PMCID: PMC5356914 DOI: 10.18632/oncotarget.12554] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 09/29/2016] [Indexed: 12/25/2022] Open
Abstract
Despite aggressive treatment for localized cancer, prostate cancer (PC) remains a leading cause of cancer-related death for American men due to a subset of patients progressing to lethal and incurable metastatic castrate-resistant prostate cancer (CRPC). Organ-confined PC is treated by surgery or radiation with or without androgen deprivation therapy (ADT), while options for locally advanced and disseminated PC include radiation combined with ADT, or systemic treatments including chemotherapy. Progression to CRPC results from failure of ADT, which targets the androgen receptor (AR) signaling axis and inhibits AR-driven proliferation and survival pathways. The exact mechanisms underlying the transition from androgen-dependent PC to CRPC remain incompletely understood. Reactivation of AR has been shown to occur in CRPC despite depletion of circulating androgens by ADT. At the same time, the presence of AR-negative cell populations in CRPC has also been identified. While AR signaling has been proposed as the primary driver of CRPC, AR-independent signaling pathways may represent additional mechanisms underlying CRPC progression. Identification of new therapeutic strategies to target both AR-positive and AR-negative PC cell populations and, thereby, AR-driven as well as non-AR-driven PC cell growth and survival mechanisms would provide a two-pronged approach to eliminate CRPC cells with potential for synthetic lethality. In this review, we provide an overview of AR-dependent and AR-independent molecular mechanisms which drive CRPC, with special emphasis on the role of the Jak2-Stat5a/b signaling pathway in promoting castrate-resistant growth of PC through both AR-dependent and AR-independent mechanisms.
Collapse
Affiliation(s)
- David T Hoang
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Kenneth A Iczkowski
- Department of Pathology, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Deepak Kilari
- Department of Medicine, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - William See
- Department of Urology, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Marja T Nevalainen
- Department of Pathology, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Pharmacology/Toxicology, Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
43
|
Murray NP, Reyes E, Fuentealba C, Jacob O, Orellana N. Possible Role of HER-2 in the Progression of Prostate Cancer from Primary Tumor to Androgen Independence. Asian Pac J Cancer Prev 2016; 16:6615-9. [PMID: 26434884 DOI: 10.7314/apjcp.2015.16.15.6615] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The expression of HER-2 in prostate cancer has been linked to disease progression. We analysed the presence of HER-2 expression in primary tumors in men undergoing radical prostatectomy, its association with clinical and pathological findings, and its expression in secondary circulating prostate cells (CPCs) during follow up, as well as links with biochemical failure and the effects of androgen blockade. MATERIALS AND METHODS Consecutive men undergoing radical prostatectomy for histologically confirmed prostate cancer were analyzed. HER-2 expression in the primary tumor was assessed using the HercepTest®, CPCs were identified from blood samples using standard immunocytochemistry with anti-PSA and positive samples with the HercepTest® to determine HER-2 expression. The influence of HER-2 expression on the frequency of biochemical failure and effects of androgen blockade was determined. RESULTS 144 men with a mean age of 64.8±10.3 years participated, with a median follow up of 8.2 years. HER-2 was expressed in 20.8% of primary tumors; it was associated with vascular infiltration and older age, but not with other clinical pathological findings. Some 40.3% of men had secondary CPCs detected, of which 38% expressed HER-2. Men CPC (+) had a higher frequency of biochemical failure, but there was no difference in HER-2 expression of CPCs with the frequency of biochemical failure. After androgen blockade, men with HER-2 (+) positive secondary CPCs had a higher frequency of disease progression to castrate resistant disease. CONCLUSIONS HER-2 plays a dual role in the progression of prostate cancer; firstly it may increase the potential of tumor cells to disseminate from the primary tumor via the blood by increasing vascular infiltration. In the presence of androgens, there is no survival advantage of expressing HER-2, but once biochemical failure has occurred and androgen blockade started, HER-2 positive cells are resistant to treatment, survive and grow leading to castration resistant disease.
Collapse
Affiliation(s)
- Nigel P Murray
- Hematology, Medicine, Hospital de Carabineros de Chile, Faculty of Medicine University Finis Terrae, Santiago, Chile E-mail :
| | | | | | | | | |
Collapse
|
44
|
Kallifatidis G, Hoy JJ, Lokeshwar BL. Bioactive natural products for chemoprevention and treatment of castration-resistant prostate cancer. Semin Cancer Biol 2016; 40-41:160-169. [PMID: 27370570 DOI: 10.1016/j.semcancer.2016.06.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 06/18/2016] [Accepted: 06/27/2016] [Indexed: 01/30/2023]
Abstract
Prostate cancer (PCa), a hormonally-driven cancer, ranks first in incidence and second in cancer related mortality in men in most Western industrialized countries. Androgen and androgen receptor (AR) are the dominant modulators of PCa growth. Over the last two decades multiple advancements in screening, treatment, surveillance and palliative care of PCa have significantly increased quality of life and survival following diagnosis. However, over 20% of patients initially diagnosed with PCa still develop an aggressive and treatment-refractory disease. Prevention or treatment for hormone-refractory PCa using bioactive compounds from marine sponges, mushrooms, and edible plants either as single agents or as adjuvants to existing therapy, has not been clinically successful. Major advancements have been made in the identification, testing and modification of the existing molecular structures of natural products. Additionally, conjugation of these compounds to novel matrices has enhanced their bio-availability; a big step towards bringing natural products to clinical trials. Natural products derived from edible plants (nutraceuticals), and common folk-medicines might offer advantages over synthetic compounds due to their broader range of targets, as compared to mostly single target synthetic anticancer compounds; e.g. kinase inhibitors. The use of synthetic inhibitors or antibodies that target a single aberrant molecule in cancer cells might be in part responsible for emergence of treatment refractory cancers. Nutraceuticals that target AR signaling (epigallocatechin gallate [EGCG], curcumin, and 5α-reductase inhibitors), AR synthesis (ericifolin, capsaicin and others) or AR degradation (betulinic acid, di-indolyl diamine, sulphoraphane, silibinin and others) are prime candidates for use as adjuvant or mono-therapies. Nutraceuticals target multiple pathophysiological mechanisms involved during cancer development and progression and thus have potential to simultaneously inhibit both prostate cancer growth and metastatic progression (e.g., inhibition of angiogenesis, epithelial-mesenchymal transition (EMT) and proliferation). Given their multi-targeting properties along with relatively lower systemic toxicity, these compounds offer significant therapeutic advantages for prevention and treatment of PCa. This review emphasizes the potential application of some of the well-researched natural compounds that target AR for prevention and therapy of PCa.
Collapse
Affiliation(s)
- Georgios Kallifatidis
- Department of Medicine, Georgia Cancer Center and Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - James J Hoy
- Department of Medicine, Georgia Cancer Center and Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Bal L Lokeshwar
- Department of Medicine, Georgia Cancer Center and Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; Biochemistry and Molecular Biology, Georgia Cancer Center and Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; Research Service, Charlie Norwood VA Hospital and Medical Center, Augusta, GA 30912, USA.
| |
Collapse
|
45
|
Wartalski K, Knet-Seweryn M, Hoja-Lukowicz D, Tabarowski Z, Duda M. Androgen receptor-mediated non-genomic effects of vinclozolin on porcine ovarian follicles and isolated granulosa cells: Vinclozolin and non-genomic effects in porcine ovarian follicles. Acta Histochem 2016; 118:377-86. [PMID: 27094116 DOI: 10.1016/j.acthis.2016.03.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 03/16/2016] [Accepted: 03/22/2016] [Indexed: 01/08/2023]
Abstract
The present study investigated the influence of the androgen receptor (AR) agonists testosterone (T) and dihydrotestosterone (DHT), and vinclozolin (Vnz), a fungicide with antiandrogenic activity, on non-genomic signal transduction within ovarian follicles. Porcine granulosa cells (GCs) isolated from mature follicles were cultured for 48h. For the last 24h of culture, they were exposed to T (10(-7)M), DHT (10(-7)M), Vnz (1.4×10(-5)M), T and Vnz (T+Vnz), or DHT and Vnz (DHT+Vnz) at the same concentrations. To better imitate in vivo conditions, whole follicles (4-6mm in diameter) were incubated (24h) in an organ culture system with the same factors. Expression of AR mRNA and protein was determined by real-time PCR and western blot analyses. To demonstrate AR localization in cultured GCs and whole follicles, immunocytochemistry and immunohistochemistry were performed, respectively. To elucidate the possible non-genomic action of Vnz in GCs, protein expression and the activity of ERK1/2 and Akt kinases were determined by western blot and ELISA analyses. The immunocytochemistry and immunohistochemistry results showed that exposure of GCs and follicles to Vnz resulted in cytoplasmic and perinuclear AR localization. Real-time PCR and western blot analysis showed that AR mRNA and protein expression increased (P≤0.001) in GC cultures after combined treatment with an androgen and Vnz. In whole follicles, such treatment also increased AR mRNA with a decrease in the respective protein expression (P≤0.001). Moreover, addition of T or DHT with Vnz increased the activity of ERK1/2 and Akt kinases in cultured GCs (P≤0.001). The results suggest a novel mechanism for Vnz action in porcine ovarian follicles on both AR mRNA and protein levels. Thus, this environmental antiandrogen activates non-genomic signaling pathways, as indicated by the increased activity of both investigated kinases observed within minutes of Vnz addition. Given the widespread presence of Vnz in the environment, elucidation of its non-genomic action should be the subject of studies on female fertility.
Collapse
|
46
|
Graham L, Schweizer MT. Targeting persistent androgen receptor signaling in castration-resistant prostate cancer. Med Oncol 2016; 33:44. [DOI: 10.1007/s12032-016-0759-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 03/24/2016] [Indexed: 12/19/2022]
|
47
|
Gao S, Ye H, Gerrin S, Wang H, Sharma A, Chen S, Patnaik A, Sowalsky AG, Voznesensky O, Han W, Yu Z, Mostaghel EA, Nelson PS, Taplin ME, Balk SP, Cai C. ErbB2 Signaling Increases Androgen Receptor Expression in Abiraterone-Resistant Prostate Cancer. Clin Cancer Res 2016; 22:3672-82. [PMID: 26936914 DOI: 10.1158/1078-0432.ccr-15-2309] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 02/13/2016] [Indexed: 02/01/2023]
Abstract
PURPOSE ErbB2 signaling appears to be increased and may enhance androgen receptor (AR) activity in a subset of patients with castration-resistant prostate cancer (CRPC), but agents targeting ErbB2 have not been effective. This study was undertaken to assess ErbB2 activity in abiraterone-resistant prostate cancer and to determine whether it may contribute to AR signaling in these tumors. EXPERIMENTAL DESIGN AR activity and ErbB2 signaling were examined in the radical prostatectomy specimens from a neoadjuvant clinical trial of leuprolide plus abiraterone and in the specimens from abiraterone-resistant CRPC xenograft models. The effect of ErbB2 signaling on AR activity was determined in two CRPC cell lines. Moreover, the effect of combination treatment with abiraterone and an ErbB2 inhibitor was assessed in a CRPC xenograft model. RESULTS We found that ErbB2 signaling was elevated in residual tumor following abiraterone treatment in a subset of patients and was associated with higher nuclear AR expression. In xenograft models, we similarly demonstrated that ErbB2 signaling was increased and associated with AR reactivation in abiraterone-resistant tumors. Mechanistically, we show that ErbB2 signaling and subsequent activation of the PI3K/AKT signaling stabilizes AR protein. Furthermore, concomitantly treating CRPC cells with abiraterone and an ErbB2 inhibitor, lapatinib, blocked AR reactivation and suppressed tumor progression. CONCLUSIONS ErbB2 signaling is elevated in a subset of patients with abiraterone-resistant prostate cancer and stabilizes AR protein. Combination therapy with abiraterone and ErbB2 antagonists may be effective for treating the subset of CRPC with elevated ErbB2 activity. Clin Cancer Res; 22(14); 3672-82. ©2016 AACR.
Collapse
MESH Headings
- Androgens/genetics
- Androstenes/pharmacology
- Animals
- Cell Line, Tumor
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/genetics
- Humans
- Leuprolide/pharmacology
- Male
- Mice
- Mice, SCID
- Phosphatidylinositol 3-Kinases/genetics
- Prostate/drug effects
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/genetics
- Receptor, ErbB-2/genetics
- Receptors, Androgen/genetics
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Xenograft Model Antitumor Assays/methods
Collapse
Affiliation(s)
- Shuai Gao
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, Massachusetts. Hematology-Oncology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Huihui Ye
- Hematology-Oncology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Sean Gerrin
- Hematology-Oncology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Hongyun Wang
- Hematology-Oncology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Ankur Sharma
- Hematology-Oncology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Sen Chen
- Hematology-Oncology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Akash Patnaik
- Hematology-Oncology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts. Department of Medicine, University of Chicago, Chicago, Illinois
| | - Adam G Sowalsky
- Hematology-Oncology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Olga Voznesensky
- Hematology-Oncology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Wanting Han
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, Massachusetts
| | - Ziyang Yu
- Hematology-Oncology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Elahe A Mostaghel
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Peter S Nelson
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Mary-Ellen Taplin
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Steven P Balk
- Hematology-Oncology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.
| | - Changmeng Cai
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, Massachusetts. Hematology-Oncology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
48
|
Ciccarese C, Santoni M, Brunelli M, Buti S, Modena A, Nabissi M, Artibani W, Martignoni G, Montironi R, Tortora G, Massari F. AR-V7 and prostate cancer: The watershed for treatment selection? Cancer Treat Rev 2015; 43:27-35. [PMID: 26827690 DOI: 10.1016/j.ctrv.2015.12.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Revised: 12/06/2015] [Accepted: 12/11/2015] [Indexed: 12/20/2022]
Abstract
The androgen receptor (AR) plays a key role in progression to metastatic castration-resistant prostate cancer (mCRPC). Despite the recent progress in targeting persistent AR activity with the next-generation hormonal therapies (abiraterone acetate and enzalutamide), resistance to these agents limits therapeutic efficacy for many patients. Several explanations for response and/or resistance to abiraterone acetate and enzalutamide are emerging, but growing interest is focusing on importance of AR splice variants (AR-Vs) and in particular of AR-V7. Increasing evidences highlight the concept that variant expression could be used as a potential predictive biomarker and a therapeutic target in advanced prostate cancer. Therefore, understanding the mechanisms of treatment resistance or sensitivity can help to achieve a more effective management of mCRPC, increasing clinical outcomes and representing a promising and engaging area of prostate cancer research.
Collapse
Affiliation(s)
- Chiara Ciccarese
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy
| | - Matteo Santoni
- Medical Oncology, AOU Ospedali Riuniti, Polytechnic University of the Marche Region, Ancona, Italy
| | - Matteo Brunelli
- Department of Pathology and Diagnostic, A.O.U.I., University of Verona, Verona, Italy
| | | | - Alessandra Modena
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy
| | - Massimo Nabissi
- School of Pharmacy, Experimental Medicine Section, University of Camerino, Camerino, Italy
| | - Walter Artibani
- Urologic Clinic, Department of Oncological and Surgical Sciences, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy
| | - Guido Martignoni
- Department of Pathology and Diagnostic, A.O.U.I., University of Verona, Verona, Italy
| | - Rodolfo Montironi
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, AOU Ospedali Riuniti, Ancona, Italy
| | - Giampaolo Tortora
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy
| | - Francesco Massari
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy.
| |
Collapse
|
49
|
Shih JW, Wang LY, Hung CL, Kung HJ, Hsieh CL. Non-Coding RNAs in Castration-Resistant Prostate Cancer: Regulation of Androgen Receptor Signaling and Cancer Metabolism. Int J Mol Sci 2015; 16:28943-78. [PMID: 26690121 PMCID: PMC4691085 DOI: 10.3390/ijms161226138] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 11/17/2015] [Accepted: 11/26/2015] [Indexed: 12/19/2022] Open
Abstract
Hormone-refractory prostate cancer frequently relapses from therapy and inevitably progresses to a bone-metastatic status with no cure. Understanding of the molecular mechanisms conferring resistance to androgen deprivation therapy has the potential to lead to the discovery of novel therapeutic targets for type of prostate cancer with poor prognosis. Progression to castration-resistant prostate cancer (CRPC) is characterized by aberrant androgen receptor (AR) expression and persistent AR signaling activity. Alterations in metabolic activity regulated by oncogenic pathways, such as c-Myc, were found to promote prostate cancer growth during the development of CRPC. Non-coding RNAs represent a diverse family of regulatory transcripts that drive tumorigenesis of prostate cancer and various other cancers by their hyperactivity or diminished function. A number of studies have examined differentially expressed non-coding RNAs in each stage of prostate cancer. Herein, we highlight the emerging impacts of microRNAs and long non-coding RNAs linked to reactivation of the AR signaling axis and reprogramming of the cellular metabolism in prostate cancer. The translational implications of non-coding RNA research for developing new biomarkers and therapeutic strategies for CRPC are also discussed.
Collapse
Affiliation(s)
- Jing-Wen Shih
- Integrated Translational Lab, The Center of Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
| | - Ling-Yu Wang
- Department of Biochemistry and Molecular Medicine, Comprehensive Cancer Center, University of California at Davis, Sacramento, CA 95817, USA.
| | - Chiu-Lien Hung
- Department of Biochemistry and Molecular Medicine, Comprehensive Cancer Center, University of California at Davis, Sacramento, CA 95817, USA.
| | - Hsing-Jien Kung
- Integrated Translational Lab, The Center of Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Biochemistry and Molecular Medicine, Comprehensive Cancer Center, University of California at Davis, Sacramento, CA 95817, USA.
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli County 35053, Taiwan.
| | - Chia-Ling Hsieh
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
50
|
Feitelson MA, Arzumanyan A, Kulathinal RJ, Blain SW, Holcombe RF, Mahajna J, Marino M, Martinez-Chantar ML, Nawroth R, Sanchez-Garcia I, Sharma D, Saxena NK, Singh N, Vlachostergios PJ, Guo S, Honoki K, Fujii H, Georgakilas AG, Bilsland A, Amedei A, Niccolai E, Amin A, Ashraf SS, Boosani CS, Guha G, Ciriolo MR, Aquilano K, Chen S, Mohammed SI, Azmi AS, Bhakta D, Halicka D, Keith WN, Nowsheen S. Sustained proliferation in cancer: Mechanisms and novel therapeutic targets. Semin Cancer Biol 2015; 35 Suppl:S25-S54. [PMID: 25892662 PMCID: PMC4898971 DOI: 10.1016/j.semcancer.2015.02.006] [Citation(s) in RCA: 464] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 02/20/2015] [Accepted: 02/23/2015] [Indexed: 02/08/2023]
Abstract
Proliferation is an important part of cancer development and progression. This is manifest by altered expression and/or activity of cell cycle related proteins. Constitutive activation of many signal transduction pathways also stimulates cell growth. Early steps in tumor development are associated with a fibrogenic response and the development of a hypoxic environment which favors the survival and proliferation of cancer stem cells. Part of the survival strategy of cancer stem cells may manifested by alterations in cell metabolism. Once tumors appear, growth and metastasis may be supported by overproduction of appropriate hormones (in hormonally dependent cancers), by promoting angiogenesis, by undergoing epithelial to mesenchymal transition, by triggering autophagy, and by taking cues from surrounding stromal cells. A number of natural compounds (e.g., curcumin, resveratrol, indole-3-carbinol, brassinin, sulforaphane, epigallocatechin-3-gallate, genistein, ellagitannins, lycopene and quercetin) have been found to inhibit one or more pathways that contribute to proliferation (e.g., hypoxia inducible factor 1, nuclear factor kappa B, phosphoinositide 3 kinase/Akt, insulin-like growth factor receptor 1, Wnt, cell cycle associated proteins, as well as androgen and estrogen receptor signaling). These data, in combination with bioinformatics analyses, will be very important for identifying signaling pathways and molecular targets that may provide early diagnostic markers and/or critical targets for the development of new drugs or drug combinations that block tumor formation and progression.
Collapse
Affiliation(s)
- Mark A Feitelson
- Department of Biology, Temple University, Philadelphia, PA, United States.
| | - Alla Arzumanyan
- Department of Biology, Temple University, Philadelphia, PA, United States
| | - Rob J Kulathinal
- Department of Biology, Temple University, Philadelphia, PA, United States
| | - Stacy W Blain
- Department of Pediatrics, State University of New York, Downstate Medical Center, Brooklyn, NY, United States
| | - Randall F Holcombe
- Tisch Cancer Institute, Mount Sinai School of Medicine, New York, NY, United States
| | - Jamal Mahajna
- MIGAL-Galilee Technology Center, Cancer Drug Discovery Program, Kiryat Shmona, Israel
| | - Maria Marino
- Department of Science, University Roma Tre, V.le G. Marconi, 446, 00146 Rome, Italy
| | - Maria L Martinez-Chantar
- Metabolomic Unit, CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Technology Park of Bizkaia, Bizkaia, Spain
| | - Roman Nawroth
- Department of Urology, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | - Isidro Sanchez-Garcia
- Experimental Therapeutics and Translational Oncology Program, Instituto de Biología Molecular y Celular del Cáncer, CSIC/Universidad de Salamanca, Salamanca, Spain
| | - Dipali Sharma
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Neeraj K Saxena
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, United States
| | - Neetu Singh
- Tissue and Cell Culture Unit, CSIR-Central Drug Research Institute, Council of Scientific & Industrial Research, Lucknow, India
| | | | - Shanchun Guo
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, Kashihara 634-8521, Japan
| | - Hiromasa Fujii
- Department of Orthopedic Surgery, Nara Medical University, Kashihara 634-8521, Japan
| | - Alexandros G Georgakilas
- Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens, Zografou 15780, Athens, Greece
| | - Alan Bilsland
- Institute of Cancer Sciences, University of Glasgow, UK
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Amr Amin
- Department of Biology, College of Science, UAE University, Al-Ain, United Arab Emirates
| | - S Salman Ashraf
- Department of Chemistry, College of Science, UAE University, Al-Ain, United Arab Emirates
| | - Chandra S Boosani
- Department of BioMedical Sciences, Creighton University, Omaha, NE, United States
| | - Gunjan Guha
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | - Maria Rosa Ciriolo
- Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Sophie Chen
- Department of Research and Development, Ovarian and Prostate Cancer Research Trust Laboratory, Guildford, Surrey GU2 7YG, United Kingdom
| | - Sulma I Mohammed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, West Lafayette, IN, United States
| | - Asfar S Azmi
- Department of Pathology, Karmonas Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Dipita Bhakta
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | - Dorota Halicka
- Brander Cancer Research Institute, Department of Pathology, New York Medical College, Valhalla, NY, United States
| | - W Nicol Keith
- Institute of Cancer Sciences, University of Glasgow, UK
| | - Somaira Nowsheen
- Mayo Graduate School, Mayo Medical School, Mayo Clinic Medical Scientist Training Program, Rochester, MN, United States
| |
Collapse
|