1
|
Zeng M, Hu Y, Zhao L, Duan C, Wu H, Xu Y, Liu X, Wang Y, Jiang D, Zeng S. Design, synthesis, and pharmacological evaluation of triazine-based PI3K/mTOR inhibitors for the potential treatment of non-small cell lung cancer. Eur J Med Chem 2024; 284:117200. [PMID: 39733482 DOI: 10.1016/j.ejmech.2024.117200] [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: 10/14/2024] [Revised: 12/20/2024] [Accepted: 12/20/2024] [Indexed: 12/31/2024]
Abstract
Dysregulated activation of the PI3K/AKT/mTOR pathway is crucial in the development of cancer, and disrupting it could potentially lead to cancer suppression, making it a viable strategy for cancer treatment. Here, as a consecutive work of our team, we described the identification and optimization of PI3K/mTOR inhibitors based on triazine scaffold, which exhibited potent PI3K/mTOR inhibitor activity. The systematically structure-activity relationship (SAR) results demonstrated that compound 5nh displayed high efficacy against PI3Kα and mTOR, with the IC50 values of 0.45 nM and 2.9 nM, respectively. Importantly, compared to the lead compound PKI-587, 5nh demonstrated significant inhibitory activity against non-small-cell lung cancer (NSCLC) cell lines, particularly HCC-827, with a 43-fold increase (3.5 nM vs 150 nM). Additionally, the compound showed effective inhibition against the EGFR-resistant variant HCC-827(GR) cell line. Mechanism validation demonstrated that 5nh significantly interfered with the PI3K/AKT/mTOR signaling pathway in HCC-827 cells. Furthermore, the oral pharmacokinetic properties of 5nh had been observably improved, with AUC0-t and Cmax increasing by 13-16 times at a dose of 10 mg/kg in mice. Importantly, the in vivo efficacy study demonstrated that orally treatment of 5nh led to significant tumor growth suppression, with a TGI value of 84.4 %. Collectively, our systematically medicinal chemistry campaigns suggested that 5nh, a novel oral available triazine derivative, held promise as a candidate for therapy of NSCLC by targeting the PI3K/AKT/mTOR cascade.
Collapse
Affiliation(s)
- Ming Zeng
- School of Pharmacy and Life Science, Jiujiang University, Jiujiang, 332005, China.
| | - Yingxuan Hu
- School of Pharmacy and Life Science, Jiujiang University, Jiujiang, 332005, China
| | - Lan Zhao
- School of Pharmacy and Life Science, Jiujiang University, Jiujiang, 332005, China
| | - Chengze Duan
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310053, China
| | - Haifeng Wu
- Zhejiang Research Institute of Chemical Industry, Hangzhou, 310023, China
| | - Yi Xu
- School of Pharmacy and Life Science, Jiujiang University, Jiujiang, 332005, China
| | - Xiaoguang Liu
- School of Pharmacy and Life Science, Jiujiang University, Jiujiang, 332005, China
| | - Yali Wang
- School of Pharmacy and Life Science, Jiujiang University, Jiujiang, 332005, China
| | - Dengzhao Jiang
- School of Pharmacy and Life Science, Jiujiang University, Jiujiang, 332005, China
| | - Shenxin Zeng
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310053, China; School of Pharmacy, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
2
|
Lee S, Lee DY, So I, Chun JN, Jeon JH. Chromatin accessibility is associated with therapeutic response in prostate cancer. Oncol Lett 2024; 28:605. [PMID: 39483964 PMCID: PMC11525612 DOI: 10.3892/ol.2024.14738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 09/12/2024] [Indexed: 11/03/2024] Open
Abstract
Treatment of advanced prostate cancer is challenging due to a lack of effective therapies. Therefore, it is important to understand the molecular mechanisms underlying therapeutic resistance in prostate cancer and to identify promising drug targets offering significant clinical advantages. Given the pivotal role of dysregulated transcriptional programs in the therapeutic response, it is essential to prioritize translational efforts targeting cancer-associated transcription factors (TFs). The present study investigated whether chromatin accessibility was associated with therapeutic resistance in prostate cancer using Assay for Transposase-Accessible Chromatin with sequencing (ATAC-seq) data. The bioinformatics analysis identified differences in chromatin accessibility between the drug response (Remission) and drug resistance (Disease) groups. Additionally, a significant association was observed between chromatin accessibility, transcriptional output and TF activity. Among TFs, forkhead box protein M1 (FOXM1) was identified as a TF with high activity and expression in the Disease group. Notably, the results of the computational analysis were validated by FOXM1 knockdown experiments, which resulted in suppressed cell proliferation and enhanced therapeutic sensitivity in prostate cancer cells. The present findings demonstrated that chromatin accessibility and TF activity may be associated with therapeutic resistance in prostate cancer. Additionally, these results provide the basis for future investigations aimed at understanding the molecular mechanisms of drug resistance and developing novel therapeutic approaches for prostate cancer.
Collapse
Affiliation(s)
- Sanghoon Lee
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Da Young Lee
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Insuk So
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Republic of Korea
| | - Jung Nyeo Chun
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Republic of Korea
| | - Ju-Hong Jeon
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Republic of Korea
| |
Collapse
|
3
|
Agbana S, McIlroy M. Extra-nuclear and cytoplasmic steroid receptor signalling in hormone dependent cancers. J Steroid Biochem Mol Biol 2024; 243:106559. [PMID: 38823459 DOI: 10.1016/j.jsbmb.2024.106559] [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: 11/09/2023] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/03/2024]
Abstract
Steroid hormone receptors are key mediators in the execution of hormone action through a combination of genomic and non-genomic action. Since their isolation and characterisation in the early 20th Century much of our understanding of the biological actions of steroid hormones are underpinned by their activated receptor activity. Over the past two decades there has been an acceleration of more omics-based research which has resulted in a major uptick in our comprehension of genomic steroid action. However, it is well understood that steroid hormones can induce very rapid signalling events in tandem with their genomic actions wherein they exert their influence through alterations in gene expression. Thus the totality of genomic and non-genomic steroid action occurs in a simultaneous and reciprocal manner and a greater appreciation of whole cell action is required to fully evaluate steroid hormone activity in vivo. In this mini-review we outline the most recent developments in non-genomic steroid action and cytoplasmic steroid hormone receptor biology in endocrine-related cancers with a focus on the 3-keto steroid receptors, in particular the androgen receptor.
Collapse
Affiliation(s)
- Stephanie Agbana
- Androgens in Health and Disease research group, RCSI University of Medicine and Health Sciences, Dublin, Ireland; Department of Surgery, RCSI University of Medicine and Health Sciences, Ireland
| | - Marie McIlroy
- Androgens in Health and Disease research group, RCSI University of Medicine and Health Sciences, Dublin, Ireland; Department of Surgery, RCSI University of Medicine and Health Sciences, Ireland.
| |
Collapse
|
4
|
De SK. New 1 H-pyrazolo[3,4- d]pyrimidin-1-yl)ethyl)-2-phenylisoquinoline-1(2 H)-ones as Phosphoinositide 3-kinase Inhibitors for Treating Cancer and Other Diseases. Recent Pat Anticancer Drug Discov 2024; 19:253-255. [PMID: 36852816 DOI: 10.2174/1574892818666230228153103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 12/20/2022] [Accepted: 01/12/2023] [Indexed: 03/01/2023]
Abstract
The patent describes novel useful compounds, such as PI3K protein kinase inhibitors, in particular as PI3K delta (δ) and/or gamma (γ) protein kinase modulators. The present disclosure also provides methods for preparing PI3K protein kinase inhibitors, pharmaceutical compositions containing them, and methods of treatment, prevention, and amelioration of PI3K kinase-mediated diseases, and disorders.
Collapse
Affiliation(s)
- Surya K De
- Department of Chemistry, Institute of Conju-Probe, San Diego, California, USA
- Department of Chemistry, Bharath University, Chennai, Tamil Nadu, 600126, India
| |
Collapse
|
5
|
Sousa A, Coelho P, Leite F, Teixeira C, Rocha AC, Santos I, Baylina P, Fernandes R, Soares R, Costa R, Gomes A. Impact of umbilical cord mesenchymal stromal/stem cell secretome and cord blood serum in prostate cancer progression. Hum Cell 2023; 36:1160-1172. [PMID: 36806993 PMCID: PMC10110723 DOI: 10.1007/s13577-023-00880-z] [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: 05/20/2022] [Accepted: 02/06/2023] [Indexed: 02/22/2023]
Abstract
Prostate cancer (PCa) is the second most common malignancy in men, and the fifth leading cause of death worldwide. Mesenchymal stromal/stem cells (MSC) have been identified in PCa, although contradictory effects in malignant transformation and tumor progression have been described. Since umbilical cord (UC) MSC and cord blood serum (CBS) are rich in numerous growth and anti-inflammatory factors, UC-MSC secretome and CBS are able to modulate tumor cell proliferation and survival as well as immunity and angiogenesis. In the present study, we address this relationship and investigate the influence of UC-MSC secretome and CBS on two human PCa cell lines (PC3 and LNCaP) and a normal epithelial prostate cell line (HPEpiC). Our results disclosed that upon exposure to UC-MSC-conditioned medium or CBS, both PC3 and LNCaP cells exhibited reduced viability, proliferation, and motility while non-malignant epithelial prostate cells were unaffected. These findings were corroborated by expression analysis of AKT/PI3K signaling pathway, p53 and interleukin genes. UC-MSC and CBS factors decreased the expression of growth-stimulating AKT and PI3K effectors and simultaneously up-regulated the expression of tumor-suppressor p53. Moreover, a more anti-inflammatory expression profile was found in both malignant PCa cell lines. Altogether, these results shed light into possible mechanisms by which UC-MSC and CBS reduce PCa progression, further reinforcing their potential use as novel therapeutic agents in PCa.
Collapse
Affiliation(s)
- André Sousa
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, Universidade do Porto, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,LaBMI - Laboratório de Biotecnologia Médica e Industrial, PORTIC - Porto Research, Technology, and Innovation Center, Polytechnic of Porto, Porto, Portugal
| | - Pedro Coelho
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, Universidade do Porto, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,School of Health (ESS), Polytechnic of Porto, Porto, Portugal
| | - Fernanda Leite
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Department of Clinical Haematology, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Catarina Teixeira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,LaBMI - Laboratório de Biotecnologia Médica e Industrial, PORTIC - Porto Research, Technology, and Innovation Center, Polytechnic of Porto, Porto, Portugal
| | - Ana Catarina Rocha
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, Universidade do Porto, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,LaBMI - Laboratório de Biotecnologia Médica e Industrial, PORTIC - Porto Research, Technology, and Innovation Center, Polytechnic of Porto, Porto, Portugal
| | - Inês Santos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,LaBMI - Laboratório de Biotecnologia Médica e Industrial, PORTIC - Porto Research, Technology, and Innovation Center, Polytechnic of Porto, Porto, Portugal
| | - Pilar Baylina
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,LaBMI - Laboratório de Biotecnologia Médica e Industrial, PORTIC - Porto Research, Technology, and Innovation Center, Polytechnic of Porto, Porto, Portugal.,School of Health (ESS), Polytechnic of Porto, Porto, Portugal
| | - Ruben Fernandes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,LaBMI - Laboratório de Biotecnologia Médica e Industrial, PORTIC - Porto Research, Technology, and Innovation Center, Polytechnic of Porto, Porto, Portugal.,FCS - Faculty of Health Sciences, HE-UFP, Hospital Escola - Universidade Fernando Pessoa, Porto, Portugal
| | - Raquel Soares
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, Universidade do Porto, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Raquel Costa
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, Universidade do Porto, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,LaBMI - Laboratório de Biotecnologia Médica e Industrial, PORTIC - Porto Research, Technology, and Innovation Center, Polytechnic of Porto, Porto, Portugal
| | - Andreia Gomes
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, Universidade do Porto, Porto, Portugal. .,Bebé Vida, Ciências Para a Vida, S.A, Av. da França 476, 4050-367, Porto, Portugal.
| |
Collapse
|
6
|
Tucci P, Brown I, Bewick GS, Pertwee RG, Marini P. The Plant Derived 3-3'-Diindolylmethane (DIM) Behaves as CB 2 Receptor Agonist in Prostate Cancer Cellular Models. Int J Mol Sci 2023; 24:ijms24043620. [PMID: 36835033 PMCID: PMC9962283 DOI: 10.3390/ijms24043620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/30/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
3-3'-Diindolylmethane (DIM) is a biologically active dimer derived from the endogenous conversion of indole-3-carbinol (I3C), a naturally occurring glucosinolate found in many cruciferous vegetables (i.e., Brassicaceae). DIM was the first pure androgen receptor antagonist isolated from the Brassicaceae family and has been recently investigated for its potential pharmacological use in prostate cancer prevention and treatment. Interestingly, there is evidence that DIM can also interact with cannabinoid receptors. In this context, by considering the well-known involvement of the endocannabinoid system in prostate cancer, we have pharmacologically characterized the properties of DIM on both CB1 and CB2 cannabinoid receptors in two human prostate cancer cell lines: PC3 (androgen-independent/androgen receptor negative) and LNCaP (androgen-dependent). In the PC3 cell line, DIM was able to activate CB2 receptors and potentially associated apoptotic pathways. On the other hand, although DIM was also able to activate CB2 receptors in the LNCaP cell line, no apoptotic effects were observed. Our evidence confirms that DIM is a CB2 receptor ligand and, moreover, it has a potential anti-proliferative effect on androgen-independent/androgen receptor-negative prostate cancer cells.
Collapse
Affiliation(s)
- Paolo Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
- Correspondence:
| | - Iain Brown
- Division of Applied Medicine, School of Medicine and Dentistry, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Guy S. Bewick
- The Institute of Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Roger G. Pertwee
- The Institute of Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Pietro Marini
- Institute of Education in Healthcare and Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, UK
| |
Collapse
|
7
|
Bernal L, Pinzi L, Rastelli G. Identification of Promising Drug Candidates against Prostate Cancer through Computationally-Driven Drug Repurposing. Int J Mol Sci 2023; 24:ijms24043135. [PMID: 36834548 PMCID: PMC9964599 DOI: 10.3390/ijms24043135] [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: 12/29/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/08/2023] Open
Abstract
Prostate cancer (PC) is one of the most common types of cancer in males. Although early stages of PC are generally associated with favorable outcomes, advanced phases of the disease present a significantly poorer prognosis. Moreover, currently available therapeutic options for the treatment of PC are still limited, being mainly focused on androgen deprivation therapies and being characterized by low efficacy in patients. As a consequence, there is a pressing need to identify alternative and more effective therapeutics. In this study, we performed large-scale 2D and 3D similarity analyses between compounds reported in the DrugBank database and ChEMBL molecules with reported anti-proliferative activity on various PC cell lines. The analyses included also the identification of biological targets of ligands with potent activity on PC cells, as well as investigations on the activity annotations and clinical data associated with the more relevant compounds emerging from the ligand-based similarity results. The results led to the prioritization of a set of drugs and/or clinically tested candidates potentially useful in drug repurposing against PC.
Collapse
Affiliation(s)
- Leonardo Bernal
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125 Modena, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Luca Pinzi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125 Modena, Italy
| | - Giulio Rastelli
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125 Modena, Italy
- Correspondence: ; Tel.: +39-059-2058564
| |
Collapse
|
8
|
Kobayashi H, Kosaka T, Nakamura K, Kimura T, Nishihara H, Oya M. Genomic analysis of aggressive ductal adenocarcinoma of the prostate. Cancer Med 2022; 12:8445-8451. [PMID: 36573306 PMCID: PMC10134333 DOI: 10.1002/cam4.5573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Genomic profile analysis using next-generation sequencing can potentially elucidate the pathogenesis of rare cancers. Ductal adenocarcinoma, a rare subtype of prostate cancer, has an aggressive nature. This is the first study to analyze the genomic profile of ductal adenocarcinoma in an Asian population. METHODS We identified 12 patients newly diagnosed with ductal adenocarcinoma of the prostate at two hospitals, and nine patients (75.0%) had the pure type. Genomic assessment was performed using either the PleSSision testing platform or FoundationOne CDx. RESULTS At least one genomic alteration occurred in 11 patients (91.7%), and the most frequently mutated gene was tumor suppressor protein p53 (TP53), which was found in six cases (50.0%). Alterations characteristic of this cohort were found in four cases (33.3%) of retinoblastoma transcriptional corepressor 1 (RB1), which was only observed in the pure type. Compared to previous study results, the frequency of genetic alterations in the phosphoinositide 3-kinase (PI3K) pathway (n = 3; 25.0%) and Wnt-β-catenin pathway (n = 5; 41.7%) was comparable, but no alterations in the DNA damage repair (DDR) pathway were observed. None of the patients presented high tumor mutation burden or microsatellite instability. CONCLUSIONS We found that the Asian cohort with ductal adenocarcinoma had actionable alterations, and a high frequency of alterations in TP53 and RB1 reflected the aggressive nature of the tumor. Genetic analysis using next-generation sequencing is expected to help elucidate the pathogenesis of ductal adenocarcinoma.
Collapse
Affiliation(s)
- Hiroaki Kobayashi
- Department of Urology Keio University School of Medicine Tokyo Japan
- Department of Urology Saiseikai Yokohamashi Tobu Hospital Kanagawa Japan
| | - Takeo Kosaka
- Department of Urology Keio University School of Medicine Tokyo Japan
| | - Kohei Nakamura
- Genomics Unit, Keio Cancer Center Keio University School of Medicine Tokyo Japan
| | - Tokuhiro Kimura
- Division of Diagnostic Pathology Saiseikai Yokohamashi Tobu Hospital Kanagawa Japan
| | - Hiroshi Nishihara
- Genomics Unit, Keio Cancer Center Keio University School of Medicine Tokyo Japan
| | - Mototsugu Oya
- Department of Urology Keio University School of Medicine Tokyo Japan
| |
Collapse
|
9
|
Fernandes S, Meyer ST, Shah JP, Adhikari AA, Kerr WG, Chisholm JD. N1-Benzyl Tryptamine Pan-SHIP1/2 Inhibitors: Synthesis and Preliminary Biological Evaluation as Anti-Tumor Agents. Molecules 2022; 27:8451. [PMID: 36500543 PMCID: PMC9738565 DOI: 10.3390/molecules27238451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/11/2022] Open
Abstract
Inhibition of phosphatidylinositol 3,4,5-trisphosphate 5-phosphatase (SHIP) with small molecule inhibitors leads to apoptosis in tumor cells. Inhibitors that target both SHIP1 and SHIP2 (pan-SHIP1/2 inhibitors) may have benefits in these areas since paralog compensation is not possible when both SHIP paralogs are being inhibited. A series of tryptamine-based pan-SHIP1/2 inhibitors have been synthesized and evaluated for their ability to inhibit the SHIP paralogs. The most active compounds were also evaluated for their effects on cancer cell lines.
Collapse
Affiliation(s)
- Sandra Fernandes
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Shea T. Meyer
- Department of Chemistry, Syracuse University, Syracuse, NY 13244, USA
| | - Jigisha P. Shah
- Department of Chemistry, Syracuse University, Syracuse, NY 13244, USA
| | | | - William G. Kerr
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - John D. Chisholm
- Department of Chemistry, Syracuse University, Syracuse, NY 13244, USA
| |
Collapse
|
10
|
Hou Y, Zhang F, Min W, Yuan K, Kuang W, Wang X, Zhu Y, Sun C, Xia F, Wang Y, Zhang H, Wang L, Yang P. Discovery of Novel Phosphoinositide-3-Kinase α Inhibitors with High Selectivity, Excellent Bioavailability, and Long-Acting Efficacy for Gastric Cancer. J Med Chem 2022; 65:9873-9892. [PMID: 35834807 DOI: 10.1021/acs.jmedchem.2c00549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Phosphoinositide-3-kinase (PI3K) overexpressed in many tumors is a promising target for cancer therapy. However, due to toxicity from the ubiquitous expression of PI3K in many tissues, the development of PI3K inhibitors with high selectivity and low toxicity has become an urgent need for tumor treatment. Herein, based on the HipHop, we designed and synthesized a series of 6-(4,6-dimorpholino-1,3,5-triazin-2-yl)benzo[d]oxazol-2-amine derivatives as potent, selective, and long-acting PI3Kα inhibitors. Compound 27 was determined with potent PI3Kα inhibitory activity (IC50 = 4.4 nM), which exhibited excellent selectivity for homologous PI3K enzymes and a 370 kinome panel. Meanwhile, 27 featured favorable stability (T1/2 > 10 h) and high bioavailability (130%). Importantly, compound 27 exerted great antigastric cancer activity in vivo when combined with taxol. Collectively, these characteristics suggested 27 to be a promising PI3K agent for cancer treatment.
Collapse
Affiliation(s)
- Yi Hou
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Fang Zhang
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Wenjian Min
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Kai Yuan
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Wenbin Kuang
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao Wang
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yasheng Zhu
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Chengliang Sun
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Fei Xia
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yanyin Wang
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Haolin Zhang
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Linping Wang
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Peng Yang
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
11
|
Dungan OM, Dormann S, Fernandes S, Duffy BC, Effiong DG, Kerr WG, Chisholm JD. Synthetic studies on the indane SHIP1 agonist AQX-1125. Org Biomol Chem 2022; 20:4016-4020. [PMID: 35506893 DOI: 10.1039/d2ob00555g] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
AQX-1125 is an indane based SHIP1 agonist that has been evaluated in the clinic for the treatment of bladder pain syndrome/interstitial cystitis. To support our own studies on SHIP1 agonists as potential treatments for IBD and Crohn's disease, a new synthetic route to the SHIP1 agonist AQX-1125 has been developed. This sequence utilizes a hydroxy-acid intermediate which allows for ready differentiation of the C6 and C7 positions. The role of the C17 alkene in the biological activity of the system is also investigated, and this functional group is not required for SHIP1 agonist activity. While AQX-1125 shows SHIP1 agonist activity in enzyme assays, it does not show activity in cell based assays similar to other SHIP1 agonists, which limits the utility of this molecule.
Collapse
Affiliation(s)
- Otto M Dungan
- Department of Chemistry, Syracuse University, 1-014 Center for Science and Technology, Syracuse, NY 13244, USA.
| | - Shawn Dormann
- Department of Chemistry, Syracuse University, 1-014 Center for Science and Technology, Syracuse, NY 13244, USA.
| | - Sandra Fernandes
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Brian C Duffy
- Department of Chemistry, Syracuse University, 1-014 Center for Science and Technology, Syracuse, NY 13244, USA.
| | - Daniel G Effiong
- Department of Chemistry, Syracuse University, 1-014 Center for Science and Technology, Syracuse, NY 13244, USA.
| | - William G Kerr
- Department of Chemistry, Syracuse University, 1-014 Center for Science and Technology, Syracuse, NY 13244, USA. .,Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA.,Department of Pediatrics, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - John D Chisholm
- Department of Chemistry, Syracuse University, 1-014 Center for Science and Technology, Syracuse, NY 13244, USA.
| |
Collapse
|
12
|
Dietrichs D, Grimm D, Sahana J, Melnik D, Corydon TJ, Wehland M, Krüger M, Vermeesen R, Baselet B, Baatout S, Hybel TE, Kahlert S, Schulz H, Infanger M, Kopp S. Three-Dimensional Growth of Prostate Cancer Cells Exposed to Simulated Microgravity. Front Cell Dev Biol 2022; 10:841017. [PMID: 35252204 PMCID: PMC8893349 DOI: 10.3389/fcell.2022.841017] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 01/25/2022] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer metastasis has an enormous impact on the mortality of cancer patients. Factors involved in cancer progression and metastasis are known to be key players in microgravity (µg)-driven three-dimensional (3D) cancer spheroid formation. We investigated PC-3 prostate cancer cells for 30 min, 2, 4 and 24 h on the random positioning machine (RPM), a device simulating µg on Earth. After a 24 h RPM-exposure, the cells could be divided into two groups: one grew as 3D multicellular spheroids (MCS), the other one as adherent monolayer (AD). No signs of apoptosis were visible. Among others, we focused on cytokines involved in the events of metastasis and MCS formation. After 24 h of exposure, in the MCS group we measured an increase in ACTB, MSN, COL1A1, LAMA3, FN1, TIMP1, FLT1, EGFR1, IL1A, IL6, CXCL8, and HIF1A mRNA expression, and in the AD group an elevation of LAMA3, COL1A1, FN1, MMP9, VEGFA, IL6, and CXCL8 mRNAs compared to samples subjected to 1 g conditions. Significant downregulations in AD cells were detected in the mRNA levels of TUBB, KRT8, IL1B, IL7, PIK3CB, AKT1 and MTOR after 24 h. The release of collagen-1α1 and fibronectin protein in the supernatant was decreased, whereas the secretion of IL-6 was elevated in 24 h RPM samples. The secretion of IL-1α, IL-1β, IL-7, IL-2, IL-8, IL-17, TNF-α, laminin, MMP-2, TIMP-1, osteopontin and EGF was not significantly altered after 24 h compared to 1 g conditions. The release of soluble factors was significantly reduced after 2 h (IL-1α, IL-2, IL-7, IL-8, IL-17, TNF-α, collagen-1α1, MMP-2, osteopontin) and elevated after 4 h (IL-1β, IL-2, IL-6, IL-7, IL-8, TNF-α, laminin) in RPM samples. Taken together, simulated µg induced 3D growth of PC-3 cancer cells combined with a differential expression of the cytokines IL-1α, IL-1β, IL-6 and IL-8, supporting their involvement in growth and progression of prostate cancer cells.
Collapse
Affiliation(s)
- Dorothea Dietrichs
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Daniela Grimm
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University Magdeburg, Magdeburg, Germany
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University Magdeburg, Magdeburg, Germany
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- *Correspondence: Daniela Grimm,
| | | | - Daniela Melnik
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Thomas J. Corydon
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Ophthalmology, Aarhus University Hospital, Aarhus, Denmark
| | - Markus Wehland
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University Magdeburg, Magdeburg, Germany
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Marcus Krüger
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University Magdeburg, Magdeburg, Germany
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Randy Vermeesen
- Radiobiology Unit, SCK CEN, Belgian Nuclear Research Centre, Mol, Belgium
| | - Bjorn Baselet
- Radiobiology Unit, SCK CEN, Belgian Nuclear Research Centre, Mol, Belgium
| | - Sarah Baatout
- Radiobiology Unit, SCK CEN, Belgian Nuclear Research Centre, Mol, Belgium
- Department of Molecular Biotechnology, Ghent University, Ghent, Belgium
| | | | - Stefan Kahlert
- Institute of Anatomy, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Herbert Schulz
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University Magdeburg, Magdeburg, Germany
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Manfred Infanger
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University Magdeburg, Magdeburg, Germany
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Sascha Kopp
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University Magdeburg, Magdeburg, Germany
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University Magdeburg, Magdeburg, Germany
| |
Collapse
|
13
|
Digoxin exerts anticancer activity on human nonsmall cell lung cancer cells by blocking PI3K/Akt pathway. Biosci Rep 2021; 41:229832. [PMID: 34549269 PMCID: PMC8495431 DOI: 10.1042/bsr20211056] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 09/16/2021] [Accepted: 09/20/2021] [Indexed: 01/03/2023] Open
Abstract
Lung cancer remains the leading cause of cancer mortality because of its metastatic potential and high malignancy. The discovery of new applications for old drugs is a shortcut for cancer therapy. We recently investigated the antitumor effect of digoxin, a well-established drug for treating heart failure, against nonsmall cell lung cancer A549 and H1299 cells. Digoxin inhibited the proliferation and colony-forming ability of the two cell lines and arrested the cell cycle at the G0/G1 phase in A549 cells and the G2/M phase in H1299 cells. Mitochondria-mediated apoptosis was induced in A549 cells but not in H1299 cells after treatment with digoxin. Moreover, digoxin inhibited the migration, invasion, adhesion and epithelial–mesenchymal transition of A549 and H1299 cells. Autophagy was induced in both cell lines after treatment with digoxin, with an increase in autophagosome foci. In addition, digoxin inhibited the phosphorylation of Akt, mTOR and p70S6K, signaling molecules of the PI3K/Akt pathway that are known to be involved in tumor cell survival, proliferation, metastasis and autophagy. Our findings suggest that digoxin has the potential to be used for therapy for human nonsmall cell lung cancer, but further evidence is required.
Collapse
|
14
|
Cham J, Venkateswaran AR, Bhangoo M. Targeting the PI3K-AKT-mTOR Pathway in Castration Resistant Prostate Cancer: A Review Article. Clin Genitourin Cancer 2021; 19:563.e1-563.e7. [PMID: 34433523 DOI: 10.1016/j.clgc.2021.07.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 01/22/2023]
Abstract
BACKGROUND Prostate cancer is one of leading causes of cancer death among men worldwide. Androgen deprivation therapy is a central part of the prostate cancer treatment algorithm, however, resistance to androgen deprivation commonly leads to disease progression. Mutations in the phosphoinositide-3-kinase pathway (PI3K) have been implicated in cancer progression and the development of castration-resistance. Thus, inhibitors of this pathway and its downstream signaling partners have been studied as potential therapeutic agents to treat metastatic castration resistant prostate cancer (mCRPC). In this article, we review recent clinical results for novel targeted therapies against the PI3K-AKT-mTOR pathway. MATERIALS AND METHODS Trials included in this systemic review were identified through conference abstracts, citations in review articles, PubMed, and ClinicalTrials.gov. Trial eligibility was independent of clinical setting or sample size. RESULTS A total of 13 prospective clinical trials between 2012 and 2020 were reviewed: Two trials for pan-PI3K inhibitors, 2 trials for selective PI3K inhibitors, 4 trials for AKT inhibitors, 5 trials for mTOR inhibitors, and 1 for a combined PI3K and mTOR inhibitor. All studies were phase I or II studies with primary outcomes of either safety and tolerability or efficacy. CONCLUSION Overall, pan-PI3K inhibitors and selective-PI3K inhibitors have not demonstrated clinical efficacy and may have significant adverse effects. AKT inhibitors may have significant adverse effects, but showed some evidence of improved survival. mTORC1 inhibitors show modest efficacy and significant adverse effects.
Collapse
Affiliation(s)
- Jason Cham
- Department of Internal Medicine, Scripps Clinic/Scripps Green Hospital, San Diego, CA.
| | | | - Munveer Bhangoo
- Department of Hematology and Oncology, Scripps Clinic/Scripps Green Hospital, San Diego, CA
| |
Collapse
|
15
|
Evolving Castration Resistance and Prostate Specific Membrane Antigen Expression: Implications for Patient Management. Cancers (Basel) 2021; 13:cancers13143556. [PMID: 34298770 PMCID: PMC8307676 DOI: 10.3390/cancers13143556] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 12/11/2022] Open
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) remains an incurable disease, despite multiple novel treatment options. The role of prostate-specific membrane antigen (PSMA) in the process of mCRPC development has long been underestimated. During the last years, a new understanding of the underlying molecular mechanisms of rising PSMA expression and its association with disease progression has emerged. Accurate understanding of these complex interactions is indispensable for a precise diagnostic process and ultimately successful treatment of advanced prostate cancer. The combination of different novel therapeutics such as androgen deprivation agents, 177LU-PSMA radioligand therapy and PARP inhibitors promises a new kind of efficacy. In this review, we summarize the current knowledge about the most relevant molecular mechanisms around PSMA in mCRPC development and how they can be implemented in mCRPC management.
Collapse
|
16
|
Xie J, Kusnadi EP, Furic L, Selth LA. Regulation of mRNA Translation by Hormone Receptors in Breast and Prostate Cancer. Cancers (Basel) 2021; 13:3254. [PMID: 34209750 PMCID: PMC8268847 DOI: 10.3390/cancers13133254] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Breast and prostate cancer are the second and third leading causes of death amongst all cancer types, respectively. Pathogenesis of these malignancies is characterised by dysregulation of sex hormone signalling pathways, mediated by the estrogen receptor-α (ER) in breast cancer and androgen receptor (AR) in prostate cancer. ER and AR are transcription factors whose aberrant function drives oncogenic transcriptional programs to promote cancer growth and progression. While ER/AR are known to stimulate cell growth and survival by modulating gene transcription, emerging findings indicate that their effects in neoplasia are also mediated by dysregulation of protein synthesis (i.e., mRNA translation). This suggests that ER/AR can coordinately perturb both transcriptional and translational programs, resulting in the establishment of proteomes that promote malignancy. In this review, we will discuss relatively understudied aspects of ER and AR activity in regulating protein synthesis as well as the potential of targeting mRNA translation in breast and prostate cancer.
Collapse
Affiliation(s)
- Jianling Xie
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia
| | - Eric P Kusnadi
- Translational Prostate Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Cancer Program, Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Luc Furic
- Translational Prostate Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Cancer Program, Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Luke A Selth
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia
- Freemasons Centre for Male Health and Wellbeing, Flinders University, Bedford Park, SA 5042, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
17
|
Hishida S, Kawakami K, Fujita Y, Kato T, Takai M, Iinuma K, Nakane K, Tsuchiya T, Koie T, Miura Y, Ito M, Mizutani K. Proteomic analysis of extracellular vesicles identified PI3K pathway as a potential therapeutic target for cabazitaxel-resistant prostate cancer. Prostate 2021; 81:592-602. [PMID: 33905554 DOI: 10.1002/pros.24138] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/07/2021] [Accepted: 04/11/2021] [Indexed: 11/07/2022]
Abstract
BACKGROUND Cabazitaxel (CBZ) is now widely used for prostate cancer (PC) patients resistant to docetaxel (DOC), however, most patients eventually acquire resistance. It will, therefore, be of great benefit to discover novel therapeutic target for the resistance. We aimed to identify candidate therapeutic targets for CBZ-resistance by proteomic analysis of extracellular vesicles (EVs) isolated from serum of DOC-resistant PC patients who later developed CBZ-resistance as well as those harvested from culture medium of DOC- and CBZ-resistant PC cell lines. METHODS Using T-cell immunoglobulin domain and mucin domain-containing protein 4 (Tim4) conjugated to magnetic beads, EVs were purified from serum of PC patients with DOC-resistance that was collected before and after acquiring CBZ-resistance and conditioned medium of DOC-resistant (22Rv1DR) and CBZ-resistant (22Rv1CR) PC cell lines. Protein analysis of EVs was performed by nanoLC-MS/MS, followed by a comparative analysis of protein expression and network analysis. The cytotoxic effect of a phosphatidylinositol-3-kinase (PI3K) inhibitor, ZSTK474, was evaluated by WST-1 assay. The expression and phosphorylation of PI3K and PTEN were examined by western blot analysis. RESULTS Among differentially regulated proteins, 77 and 61 proteins were significantly increased in EVs from CBZ-resistant PC cell line and patients, respectively. A comparison between the two datasets revealed that six proteins, fructose-bisphosphate aldolase, cytosolic nonspecific dipeptidase, CD63, CD151, myosin light chain 9, and peroxiredoxin-6 were elevated in EVs from both cell line and patients. Network analysis of the increased EV proteins identified pathways associated with CBZ-resistance including PI3K signaling pathway. ZSTK474 significantly inhibited growth of 22Rv1CR cells and improved their sensitivity to CBZ. In 22Rv1CR cells, PI3K was activated and PTEN that inhibits PI3K was deactivated. CONCLUSIONS Proteomic analysis of serum EVs was successfully accomplished by using Tim-4 as a tool to isolate highly purified EVs. Our results suggest that the combination use of CBZ and PI3K inhibitor could be a promising treatment option for CBZ-resistant PC patients.
Collapse
Affiliation(s)
- Seiji Hishida
- Department of Urology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Kyojiro Kawakami
- Research Team for Mechanism of Aging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Yasunori Fujita
- Research Team for Functional Biogerontology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Taku Kato
- Department of Urology, Asahi University Hospital, Gifu, Japan
| | - Manabu Takai
- Department of Urology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Koji Iinuma
- Department of Urology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Keita Nakane
- Department of Urology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Tomohiro Tsuchiya
- Department of Urology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Takuya Koie
- Department of Urology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Yuri Miura
- Research Team for Mechanism of Aging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Masafumi Ito
- Research Team for Functional Biogerontology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Kosuke Mizutani
- Department of Urology, Gifu University Graduate School of Medicine, Gifu, Japan
| |
Collapse
|
18
|
Xue J, Chen K, Hu H, Gopinath SCB. Progress in gene therapy treatments for prostate cancer. Biotechnol Appl Biochem 2021; 69:1166-1175. [PMID: 33988271 DOI: 10.1002/bab.2193] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 05/12/2021] [Indexed: 01/17/2023]
Abstract
Prostate cancer is one of the predominant cancers affecting men and has been widely reported. In the past, various therapies and drugs have been proposed to treat prostate cancer. Among these treatments, gene therapy has been considered to be an optimal and widely applicable treatment. Furthermore, due to the increased specificity of gene sequence complementation, the targeted delivery of complementary gene sequences may represent a useful treatment in certain instances. Various gene therapies, including tumor-suppressor gene therapy, suicide gene therapy, immunomodulation gene therapy and anti-oncogene therapies, have been established to treat a wide range of diseases, such as cardiac disease, cystic fibrosis, HIV/AIDS, diabetes, hemophilia, and cancers. To this end, several gene therapy clinical trials at various phases are underway. This overview describes the developments and progress in gene therapy, with a special focus being placed on prostate cancer.
Collapse
Affiliation(s)
- Jingxin Xue
- Department of Urology, Affiliated Jinan Third Hospital of Jining Medical University, Jining Medical University, Jinan, Shandong, China
| | - Keming Chen
- Department of Urology, Affiliated Jinan Third Hospital of Jining Medical University, Jining Medical University, Jinan, Shandong, China
| | - Heyi Hu
- Department of Urology, Affiliated Jinan Third Hospital of Jining Medical University, Jining Medical University, Jinan, Shandong, China
| | - Subash C B Gopinath
- Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), Kangar, Perlis, 01000, Malaysia.,Faculty of Chemical Engineering Technology, Universiti Malaysia Perlis (UniMAP), Arau, Perlis, 02600, Malaysia
| |
Collapse
|
19
|
Ranasinghe W, Shapiro DD, Zhang M, Bathala T, Navone N, Thompson TC, Broom B, Aparicio A, Tu SM, Tang C, Davis JW, Pisters L, Chapin BF. Optimizing the diagnosis and management of ductal prostate cancer. Nat Rev Urol 2021; 18:337-358. [PMID: 33824525 DOI: 10.1038/s41585-021-00447-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2021] [Indexed: 12/13/2022]
Abstract
Ductal adenocarcinoma (DAC) is the most common variant histological subtype of prostate carcinoma and has an aggressive clinical course. DAC is usually characterized and treated as high-risk prostatic acinar adenocarcinoma (PAC). However, DAC has a different biology to that of acinar disease, which often poses a challenge for both diagnosis and management. DAC can be difficult to identify using conventional diagnostic modalities such as serum PSA levels and multiparametric MRI, and the optimal management for localized DAC is unknown owing to the rarity of the disease. Following definitive therapy for localized disease with radical prostatectomy or radiotherapy, the majority of DACs recur with visceral metastases at low PSA levels. Various systemic therapies that have been shown to be effective in high-risk PAC have limited use in treating DAC. Although current understanding of the biology of DAC is limited, genomic analyses have provided insights into the pathology behind its aggressive behaviour and potential future therapeutic targets.
Collapse
Affiliation(s)
- Weranja Ranasinghe
- Department of Urology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
| | - Daniel D Shapiro
- Department of Urology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Miao Zhang
- Department of Pathology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Tharakeswara Bathala
- Department of Radiology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Nora Navone
- Department of Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Timothy C Thompson
- Department of Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Bradley Broom
- Department of Bioinformatics and Computational Biology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Ana Aparicio
- Department of Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Shi-Ming Tu
- Department of Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Chad Tang
- Department of Radiation Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - John W Davis
- Department of Urology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Louis Pisters
- Department of Urology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Brian F Chapin
- Department of Urology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
20
|
Samaržija I. Post-Translational Modifications That Drive Prostate Cancer Progression. Biomolecules 2021; 11:247. [PMID: 33572160 PMCID: PMC7915076 DOI: 10.3390/biom11020247] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/04/2021] [Accepted: 02/06/2021] [Indexed: 02/07/2023] Open
Abstract
While a protein primary structure is determined by genetic code, its specific functional form is mostly achieved in a dynamic interplay that includes actions of many enzymes involved in post-translational modifications. This versatile repertoire is widely used by cells to direct their response to external stimuli, regulate transcription and protein localization and to keep proteostasis. Herein, post-translational modifications with evident potency to drive prostate cancer are explored. A comprehensive list of proteome-wide and single protein post-translational modifications and their involvement in phenotypic outcomes is presented. Specifically, the data on phosphorylation, glycosylation, ubiquitination, SUMOylation, acetylation, and lipidation in prostate cancer and the enzymes involved are collected. This type of knowledge is especially valuable in cases when cancer cells do not differ in the expression or mutational status of a protein, but its differential activity is regulated on the level of post-translational modifications. Since their driving roles in prostate cancer, post-translational modifications are widely studied in attempts to advance prostate cancer treatment. Current strategies that exploit the potential of post-translational modifications in prostate cancer therapy are presented.
Collapse
Affiliation(s)
- Ivana Samaržija
- Laboratory for Epigenomics, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| |
Collapse
|
21
|
Zhou W, Ding X, Jin P, Li P. miR-6838-5p Affects Cell Growth, Migration, and Invasion by Targeting GPRIN3 via the Wnt/β-Catenin Signaling Pathway in Gastric Cancer. Pathobiology 2020; 87:327-337. [PMID: 33254176 DOI: 10.1159/000511691] [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] [Received: 03/02/2020] [Accepted: 09/16/2020] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) is a highly prevalent digestive malignant tumor, ranking second in the tumor-related mortality globally. The microRNAs have been confirmed to be connected with GC progression. Accumulative evidence has suggested that miR-6838-5p exerts a suppressive effect on human cancers. Nonetheless, whether miR-6838-5p is involved in the regulation of GC remains to be investigated. During our research, miR-6838-5p was downregulated in GC cells. Upregulated miR-6838-5p repressed GC cell cycle progression, proliferation, migration, and invasion. Furthermore, miR-6838-5p overexpression repressed the nuclear import of β-catenin, thus inactivating Wnt/β-catenin pathway. Moreover, we observed that GPRIN3 was targeted by miR-6838-5p in GC with luciferase reporter and RIP assays. GPRIN3 upregulation reversed the suppression of miR-6838-5p in GC cellular processes. These findings suggest miR-6838-5p restrains the malignant behaviors of GC cells via targeting GPRIN3 to repress Wnt/β-catenin signaling pathway, which may provide novel targets for GC treatment.
Collapse
Affiliation(s)
- Weidong Zhou
- Department of Gastroenterology, Hwa Mei Hospital, University of Chinese Academy of Sciences (Ningbo No. 2 Hospital), Ningbo, China
| | - Xiaoyun Ding
- Department of Gastroenterology, Ningbo First Hospital, Ningbo, China
| | - Peihua Jin
- Department of Gastroenterology, Hwa Mei Hospital, University of Chinese Academy of Sciences (Ningbo No. 2 Hospital), Ningbo, China
| | - Peifei Li
- Department of Gastroenterology, Ningbo First Hospital, Ningbo, China,
| |
Collapse
|
22
|
Luszczak S, Simpson BS, Stopka-Farooqui U, Sathyadevan VK, Echeverria LMC, Kumar C, Costa H, Haider A, Freeman A, Jameson C, Ratynska M, Ben-Salha I, Sridhar A, Shaw G, Kelly JD, Pye H, Gately KA, Whitaker HC, Heavey S. Co-targeting PIM and PI3K/mTOR using multikinase inhibitor AUM302 and a combination of AZD-1208 and BEZ235 in prostate cancer. Sci Rep 2020; 10:14380. [PMID: 32873828 PMCID: PMC7463239 DOI: 10.1038/s41598-020-71263-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 08/13/2020] [Indexed: 12/14/2022] Open
Abstract
PIM and PI3K/mTOR pathways are often dysregulated in prostate cancer, and may lead to decreased survival, increased metastasis and invasion. The pathways are heavily interconnected and act on a variety of common effectors that can lead to the development of resistance to drug inhibitors. Most current treatments exhibit issues with toxicity and resistance. We investigated the novel multikinase PIM/PI3K/mTOR inhibitor, AUM302, versus a combination of the PIM inhibitor, AZD-1208, and the PI3K/mTOR inhibitor BEZ235 (Dactolisib) to determine their impact on mRNA and phosphoprotein expression, as well as their functional efficacy. We have determined that around 20% of prostate cancer patients overexpress the direct targets of these drugs, and this cohort are more likely to have a high Gleason grade tumour (≥ Gleason 8). A co-targeted inhibition approach offered broader inhibition of genes and phosphoproteins in the PI3K/mTOR pathway, when compared to single kinase inhibition. The preclinical inhibitor AUM302, used at a lower dose, elicited a comparable or superior functional outcome compared with combined AZD-1208 + BEZ235, which have been investigated in clinical trials, and could help to reduce treatment toxicity in future trials. We believe that a co-targeting approach is a viable therapeutic strategy that should be developed further in pre-clinical studies.
Collapse
Affiliation(s)
- Sabina Luszczak
- Molecular Diagnostics and Therapeutics Group, University College London, London, UK
| | - Benjamin S Simpson
- Molecular Diagnostics and Therapeutics Group, University College London, London, UK
| | | | | | | | - Christopher Kumar
- Molecular Diagnostics and Therapeutics Group, University College London, London, UK
| | - Helena Costa
- Research Department of Pathology, University College London, London, UK
| | - Aiman Haider
- Research Department of Pathology, University College London, London, UK
| | - Alex Freeman
- Research Department of Pathology, University College London, London, UK
| | - Charles Jameson
- Research Department of Pathology, University College London, London, UK
| | - Marzena Ratynska
- Research Department of Pathology, University College London, London, UK
| | - Imen Ben-Salha
- Research Department of Pathology, University College London, London, UK
| | - Ashwin Sridhar
- Department of Uro-Oncology, UCLH NHS Foundation Trust, London, UK
| | - Greg Shaw
- Department of Uro-Oncology, UCLH NHS Foundation Trust, London, UK
| | - John D Kelly
- Department of Uro-Oncology, UCLH NHS Foundation Trust, London, UK
| | - Hayley Pye
- Molecular Diagnostics and Therapeutics Group, University College London, London, UK
| | - Kathy A Gately
- Trinity Translational Medicine Institute, St. James's Hospital Dublin, Dublin 8, Ireland
| | - Hayley C Whitaker
- Molecular Diagnostics and Therapeutics Group, University College London, London, UK
| | - Susan Heavey
- Molecular Diagnostics and Therapeutics Group, University College London, London, UK.
| |
Collapse
|
23
|
Abd. Wahab NA, H. Lajis N, Abas F, Othman I, Naidu R. Mechanism of Anti-Cancer Activity of Curcumin on Androgen-Dependent and Androgen-Independent Prostate Cancer. Nutrients 2020; 12:E679. [PMID: 32131560 PMCID: PMC7146610 DOI: 10.3390/nu12030679] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/22/2020] [Accepted: 02/26/2020] [Indexed: 12/22/2022] Open
Abstract
Prostate cancer (PCa) is a heterogeneous disease and ranked as the second leading cause of cancer-related deaths in males worldwide. The global burden of PCa keeps rising regardless of the emerging cutting-edge technologies for treatment and drug designation. There are a number of treatment options which are effectively treating localised and androgen-dependent PCa (ADPC) through hormonal and surgery treatments. However, over time, these cancerous cells progress to androgen-independent PCa (AIPC) which continuously grow despite hormone depletion. At this particular stage, androgen depletion therapy (ADT) is no longer effective as these cancerous cells are rendered hormone-insensitive and capable of growing in the absence of androgen. AIPC is a lethal type of disease which leads to poor prognosis and is a major contributor to PCa death rates. A natural product-derived compound, curcumin has been identified as a pleiotropic compound which capable of influencing and modulating a diverse range of molecular targets and signalling pathways in order to exhibit its medicinal properties. Due to such multi-targeted behaviour, its benefits are paramount in combating a wide range of diseases including inflammation and cancer disease. Curcumin exhibits anti-cancer properties by suppressing cancer cells growth and survival, inflammation, invasion, cell proliferation as well as possesses the ability to induce apoptosis in malignant cells. In this review, we investigate the mechanism of curcumin by modulating multiple signalling pathways such as androgen receptor (AR) signalling, activating protein-1 (AP-1), phosphatidylinositol 3-kinases/the serine/threonine kinase (PI3K/Akt/mTOR), wingless (Wnt)/ß-catenin signalling, and molecular targets including nuclear factor kappa-B (NF-κB), B-cell lymphoma 2 (Bcl-2) and cyclin D1 which are implicated in the development and progression of both types of PCa, ADPC and AIPC. In addition, the role of microRNAs and clinical trials on the anti-cancer effects of curcumin in PCa patients were also reviewed.
Collapse
Affiliation(s)
- Nurul Azwa Abd. Wahab
- Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia; (N.A.A.W.); (I.O.)
| | - Nordin H. Lajis
- Laboratory of Natural Products, Faculty of Science, Universiti Putra Malaysia, UPM, Serdang 43400, Malaysia; (N.H.L.); (F.A.)
| | - Faridah Abas
- Laboratory of Natural Products, Faculty of Science, Universiti Putra Malaysia, UPM, Serdang 43400, Malaysia; (N.H.L.); (F.A.)
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, UPM, Serdang 43400, Malaysia
| | - Iekhsan Othman
- Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia; (N.A.A.W.); (I.O.)
| | - Rakesh Naidu
- Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia; (N.A.A.W.); (I.O.)
| |
Collapse
|
24
|
Small molecule targeting of SHIP1 and SHIP2. Biochem Soc Trans 2020; 48:291-300. [DOI: 10.1042/bst20190775] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/14/2020] [Accepted: 01/20/2020] [Indexed: 02/07/2023]
Abstract
Modulating the activity of the Src Homology 2 (SH2) — containing Inositol 5′-Phosphatase (SHIP) enzyme family with small molecule inhibitors provides a useful and unconventional method of influencing cell signaling in the PI3K pathway. The development of small molecules that selectively target one of the SHIP paralogs (SHIP1 or SHIP2) as well as inhibitors that simultaneously target both enzymes have provided promising data linking the phosphatase activity of the SHIP enzymes to disorders and disease states that are in dire need of new therapeutic targets. These include cancer, immunotherapy, diabetes, obesity, and Alzheimer's disease. In this mini-review, we will provide a brief overview of research in these areas that support targeting SHIP1, SHIP2 or both enzymes for therapeutic purposes.
Collapse
|
25
|
Luszczak S, Kumar C, Sathyadevan VK, Simpson BS, Gately KA, Whitaker HC, Heavey S. PIM kinase inhibition: co-targeted therapeutic approaches in prostate cancer. Signal Transduct Target Ther 2020; 5:7. [PMID: 32296034 PMCID: PMC6992635 DOI: 10.1038/s41392-020-0109-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/05/2019] [Accepted: 12/13/2019] [Indexed: 01/09/2023] Open
Abstract
PIM kinases have been shown to play a role in prostate cancer development and progression, as well as in some of the hallmarks of cancer, especially proliferation and apoptosis. Their upregulation in prostate cancer has been correlated with decreased patient overall survival and therapy resistance. Initial efforts to inhibit PIM with monotherapies have been hampered by compensatory upregulation of other pathways and drug toxicity, and as such, it has been suggested that co-targeting PIM with other treatment approaches may permit lower doses and be a more viable option in the clinic. Here, we present the rationale and basis for co-targeting PIM with inhibitors of PI3K/mTOR/AKT, JAK/STAT, MYC, stemness, and RNA Polymerase I transcription, along with other therapies, including androgen deprivation, radiotherapy, chemotherapy, and immunotherapy. Such combined approaches could potentially be used as neoadjuvant therapies, limiting the development of resistance to treatments or sensitizing cells to other therapeutics. To determine which drugs should be combined with PIM inhibitors for each patient, it will be key to develop companion diagnostics that predict response to each co-targeted option, hopefully providing a personalized medicine pathway for subsets of prostate cancer patients in the future.
Collapse
Affiliation(s)
- Sabina Luszczak
- Molecular Diagnostics and Therapeutics Group, University College London, London, UK
| | - Christopher Kumar
- Molecular Diagnostics and Therapeutics Group, University College London, London, UK
| | | | - Benjamin S Simpson
- Molecular Diagnostics and Therapeutics Group, University College London, London, UK
| | - Kathy A Gately
- Trinity Translational Medicine Institute, St. James's Hospital Dublin, Dublin 8, Dublin, Ireland
| | - Hayley C Whitaker
- Molecular Diagnostics and Therapeutics Group, University College London, London, UK
| | - Susan Heavey
- Molecular Diagnostics and Therapeutics Group, University College London, London, UK.
| |
Collapse
|
26
|
Chun JN, Cho M, Park S, So I, Jeon JH. The conflicting role of E2F1 in prostate cancer: A matter of cell context or interpretational flexibility? Biochim Biophys Acta Rev Cancer 2019; 1873:188336. [PMID: 31870703 DOI: 10.1016/j.bbcan.2019.188336] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 12/20/2019] [Indexed: 02/07/2023]
Abstract
The transcription factor E2F1 plays a crucial role in mediating multiple cancer hallmark capabilities that regulate cell cycle, survival, apoptosis, metabolism, and metastasis. Aberrant activation of E2F1 is closely associated with a poor clinical outcome in various human cancers. However, E2F1 has conflictingly been reported to exert tumor suppressive activity, raising a question as to the nature of its substantive role in the control of cell fate. In this review, we summarize deregulated E2F1 activity and its role in prostate cancer. We highlight the recent advances in understanding the molecular mechanism by which E2F1 regulates the development and progression of prostate cancer, providing insight into how cell context or data interpretation shapes the role of E2F1 in prostate cancer. This review will aid in translating biomedical knowledge into therapeutic strategies for prostate cancer.
Collapse
Affiliation(s)
- Jung Nyeo Chun
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Republic of Korea
| | - Minsoo Cho
- Undergraduate Research Program, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Soonbum Park
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Insuk So
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Republic of Korea
| | - Ju-Hong Jeon
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Republic of Korea.
| |
Collapse
|
27
|
Wegner KA, Mueller BR, Unterberger CJ, Avila EJ, Ruetten H, Turco AE, Oakes SR, Girardi NM, Halberg RB, Swanson SM, Marker PC, Vezina CM. Prostate epithelial-specific expression of activated PI3K drives stromal collagen production and accumulation. J Pathol 2019; 250:231-242. [PMID: 31674011 DOI: 10.1002/path.5363] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/30/2019] [Accepted: 10/27/2019] [Indexed: 01/01/2023]
Abstract
We genetically engineered expression of an activated form of P110 alpha, the catalytic subunit of PI3K, in mouse prostate epithelium to create a mouse model of direct PI3K activation (Pbsn-cre4Prb;PI3KGOF/+ ). We hypothesized that direct activation would cause rapid neoplasia and cancer progression. Pbsn-cre4Prb;PI3KGOF/+ mice developed widespread prostate intraepithelial hyperplasia, but stromal invasion was limited and overall progression was slower than anticipated. However, the model produced profound and progressive stromal remodeling prior to explicit epithelial neoplasia. Increased stromal cellularity and inflammatory infiltrate were evident as early as 4 months of age and progressively increased through 12 months of age, the terminal endpoint of this study. Prostatic collagen density and phosphorylated SMAD2-positive prostatic stromal cells were expansive and accumulated with age, consistent with pro-fibrotic TGF-β pathway activation. Few reported mouse models accumulate prostate-specific collagen to the degree observed in Pbsn-cre4Prb;PI3KGOF/+ . Our results indicate a signaling process beginning with prostatic epithelial PI3K and TGF-β signaling that drives prostatic stromal hypertrophy and collagen accumulation. These mice afford a unique opportunity to explore molecular mechanisms of prostatic collagen accumulation that is relevant to cancer progression, metastasis, inflammation and urinary dysfunction. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Kyle A Wegner
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, WI, USA.,University of Wisconsin-Madison/UMASS Boston George M. O'Brien Center for Benign Urologic Research, Madison, WI, USA.,Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Brett R Mueller
- University of Wisconsin-Madison/UMASS Boston George M. O'Brien Center for Benign Urologic Research, Madison, WI, USA.,Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Christopher J Unterberger
- University of Wisconsin-Madison/UMASS Boston George M. O'Brien Center for Benign Urologic Research, Madison, WI, USA.,School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Enrique J Avila
- University of Wisconsin-Madison/UMASS Boston George M. O'Brien Center for Benign Urologic Research, Madison, WI, USA.,School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Hannah Ruetten
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Anne E Turco
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, WI, USA.,University of Wisconsin-Madison/UMASS Boston George M. O'Brien Center for Benign Urologic Research, Madison, WI, USA.,Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Steven R Oakes
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA.,Department of Biomedical Engineering, College of Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Nicholas M Girardi
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Richard B Halberg
- Department of Oncology, McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, USA.,Division of Gastroenterology and Hepatology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Steven M Swanson
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Paul C Marker
- University of Wisconsin-Madison/UMASS Boston George M. O'Brien Center for Benign Urologic Research, Madison, WI, USA.,School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Chad M Vezina
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, WI, USA.,University of Wisconsin-Madison/UMASS Boston George M. O'Brien Center for Benign Urologic Research, Madison, WI, USA.,Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA.,School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
28
|
Testa U, Castelli G, Pelosi E. Cellular and Molecular Mechanisms Underlying Prostate Cancer Development: Therapeutic Implications. MEDICINES (BASEL, SWITZERLAND) 2019; 6:E82. [PMID: 31366128 PMCID: PMC6789661 DOI: 10.3390/medicines6030082] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/19/2019] [Accepted: 07/25/2019] [Indexed: 12/15/2022]
Abstract
Prostate cancer is the most frequent nonskin cancer and second most common cause of cancer-related deaths in man. Prostate cancer is a clinically heterogeneous disease with many patients exhibiting an aggressive disease with progression, metastasis, and other patients showing an indolent disease with low tendency to progression. Three stages of development of human prostate tumors have been identified: intraepithelial neoplasia, adenocarcinoma androgen-dependent, and adenocarcinoma androgen-independent or castration-resistant. Advances in molecular technologies have provided a very rapid progress in our understanding of the genomic events responsible for the initial development and progression of prostate cancer. These studies have shown that prostate cancer genome displays a relatively low mutation rate compared with other cancers and few chromosomal loss or gains. The ensemble of these molecular studies has led to suggest the existence of two main molecular groups of prostate cancers: one characterized by the presence of ERG rearrangements (~50% of prostate cancers harbor recurrent gene fusions involving ETS transcription factors, fusing the 5' untranslated region of the androgen-regulated gene TMPRSS2 to nearly the coding sequence of the ETS family transcription factor ERG) and features of chemoplexy (complex gene rearrangements developing from a coordinated and simultaneous molecular event), and a second one characterized by the absence of ERG rearrangements and by the frequent mutations in the E3 ubiquitin ligase adapter SPOP and/or deletion of CDH1, a chromatin remodeling factor, and interchromosomal rearrangements and SPOP mutations are early events during prostate cancer development. During disease progression, genomic and epigenomic abnormalities accrued and converged on prostate cancer pathways, leading to a highly heterogeneous transcriptomic landscape, characterized by a hyperactive androgen receptor signaling axis.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Vaile Regina Elena 299, 00161 Rome, Italy.
| | - Germana Castelli
- Department of Oncology, Istituto Superiore di Sanità, Vaile Regina Elena 299, 00161 Rome, Italy
| | - Elvira Pelosi
- Department of Oncology, Istituto Superiore di Sanità, Vaile Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
29
|
Dysregulated Transcriptional Control in Prostate Cancer. Int J Mol Sci 2019; 20:ijms20122883. [PMID: 31200487 PMCID: PMC6627928 DOI: 10.3390/ijms20122883] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/06/2019] [Accepted: 06/07/2019] [Indexed: 12/24/2022] Open
Abstract
Recent advances in whole-genome and transcriptome sequencing of prostate cancer at different stages indicate that a large number of mutations found in tumors are present in non-protein coding regions of the genome and lead to dysregulated gene expression. Single nucleotide variations and small mutations affecting the recruitment of transcription factor complexes to DNA regulatory elements are observed in an increasing number of cases. Genomic rearrangements may position coding regions under the novel control of regulatory elements, as exemplified by the TMPRSS2-ERG fusion and the amplified enhancer identified upstream of the androgen receptor (AR) gene. Super-enhancers are increasingly found to play important roles in aberrant oncogenic transcription. Several players involved in these processes are currently being evaluated as drug targets and may represent new vulnerabilities that can be exploited for prostate cancer treatment. They include factors involved in enhancer and super-enhancer function such as bromodomain proteins and cyclin-dependent kinases. In addition, non-coding RNAs with an important gene regulatory role are being explored. The rapid progress made in understanding the influence of the non-coding part of the genome and of transcription dysregulation in prostate cancer could pave the way for the identification of novel treatment paradigms for the benefit of patients.
Collapse
|
30
|
Bloom Syndrome Protein Activates AKT and PRAS40 in Prostate Cancer Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3685817. [PMID: 31210839 PMCID: PMC6532288 DOI: 10.1155/2019/3685817] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/07/2019] [Accepted: 03/25/2019] [Indexed: 12/16/2022]
Abstract
Purpose Prostate cancer (PC) is a common malignant tumor and a leading cause of cancer-related death in men worldwide. In order to design new therapeutic interventions for PC, an understanding of the molecular events underlying PC tumorigenesis is required. Bloom syndrome protein (BLM) is a RecQ-like helicase, which helps maintain genetic stability. BLM dysfunction has been implicated in tumor development, most recently during PC tumorigenesis. However, the molecular basis for BLM-induced PC progression remains poorly characterized. In this study, we investigated whether BLM modulates the phosphorylation of an array of prooncogenic signaling pathways to promote PC progression. Methods We analyzed differentially expressed proteins (DEPs) using iTRAQ technology. Site-directed knockout of BLM in PC-3 prostate cancer cells was performed using CRISPR/Cas9-mediated homologous recombination gene editing to confirm the effects of BLM on DEPs. PathScan® Antibody Array Kits were used to analyze the phosphorylation of nodal proteins in PC tissue. Immunohistochemistry and automated western blot (WES) analyses were used to validate these findings. Results We found that silencing BLM in PC-3 cells significantly reduced their proliferative capacity. In addition, BLM downregulation significantly reduced levels of phosphorylated protein kinase B (AKT (Ser473)) and proline-rich AKT substrate of 40 kDa (PRAS40 (Thr246)), and this was accompanied by enhanced ROS (reactive oxygen species) levels. In addition, we found that AKT and PRAS40 inhibition reduced BLM, increased ROS levels, and induced PC cell apoptosis. Conclusions We demonstrated that BLM activates AKT and PRAS40 to promote PC cell proliferation and survival. We further propose that ROS act in concert with BLM to facilitate PC oncogenesis, potentially via further enhancing AKT signaling and downregulating PTEN expression. Importantly, inhibiting the BLM-AKT-PRAS40 axis induced PC cell apoptosis. Thus, we highlight new avenues for novel anti-PC treatments.
Collapse
|