1
|
Ding Z, Lu Y, Zhao J, Zhang D, Gao B. Network Pharmacology and Molecular Dynamics Identified Potential Androgen Receptor-Targeted Metabolites in Crocus alatavicus. Int J Mol Sci 2025; 26:3533. [PMID: 40331986 PMCID: PMC12027412 DOI: 10.3390/ijms26083533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
The objective of this study is to identify the active components of Crocus alatavicus and potential targets through a combination of network pharmacology, molecular docking technology combined with molecular dynamics simulation, and binding free energy analyses. A total of 253 active ingredients from C. alatavicus were screened, and 1360 associated targets were predicted through systematic searches conducted using TCMSP, SwissDrugDesign, and SymMap, which were integrated to construct a pharmacological network to dissect the relationships among active components, targets, diseases, and pathways; we found prostate cancer-related genes were significantly enriched among the targets. Subsequently, the core prostate cancer-related targets were identified in the network, and the binding interactions between protein targets and active components were evaluated using molecular docking technology. Furthermore, molecular dynamics simulation and binding free energy analyses were performed to verify the binding stability of the most promising complex. Then, protein-protein interaction network analysis was conducted to evaluate the core target sites, leading to the identification of nine target proteins with significant correlations, providing potential targets for cancer treatment. Furthermore, these targets were found to be associated with 20 signaling pathways, including neuroactive ligand-receptor interactions, prostate cancer, lipid metabolism and atherosclerosis, as well as calcium signaling pathways. The active component-target-disease-pathway network diagram suggests that Capillarisin, Eugenol, 1-(4-Methoxyphenyl)-1-propanol, 2,4,2',4'-tetrahydroxy-3'-prenylchalcone, and 4-Hydroxymandelonitrile may serve as key components targeting prostate cancer. Molecular docking analyses demonstrated that Capillarisin has a high affinity for the androgen receptor (AR), and molecular dynamics simulation was performed to further verify the binding stability, indicating that Capillarisin may exert its pharmacological effects in prostate cancer. Based on the integrated strategies of network pharmacology, molecular docking, molecular dynamics simulation, and binding free energy analysis, this study generated novel insights into the active components of C. alatavicus and potential targets related to prostate cancer, thus providing valuable biological resources for future drug research and development.
Collapse
Affiliation(s)
- Zhen Ding
- State Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China; (Z.D.); (Y.L.); (J.Z.)
- Xinjiang Key Laboratory of Conservation and Utilization of Plant Gene Resources, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China
- University of Chinese Academy of Sciences, Beijing 100019, China
| | - Yuanfeng Lu
- State Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China; (Z.D.); (Y.L.); (J.Z.)
- Xinjiang Key Laboratory of Conservation and Utilization of Plant Gene Resources, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China
- College of Life Sciences, Nanjing Forestry University, Nanjing 210008, China
| | - Jichen Zhao
- State Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China; (Z.D.); (Y.L.); (J.Z.)
- Xinjiang Key Laboratory of Conservation and Utilization of Plant Gene Resources, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China
- University of Chinese Academy of Sciences, Beijing 100019, China
| | - Daoyuan Zhang
- State Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China; (Z.D.); (Y.L.); (J.Z.)
- Xinjiang Key Laboratory of Conservation and Utilization of Plant Gene Resources, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China
| | - Bei Gao
- State Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China; (Z.D.); (Y.L.); (J.Z.)
- Xinjiang Key Laboratory of Conservation and Utilization of Plant Gene Resources, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China
| |
Collapse
|
2
|
Bugoye FC, Torrorey-Sawe R, Biegon R, Dharsee N, Mafumiko F, Kibona H, Aboud S, Patel K, Mining S. Exploring therapeutic applications of PTEN, TMPRSS2:ERG fusion, and tumour molecular subtypes in prostate cancer management. Front Oncol 2025; 15:1521204. [PMID: 40165885 PMCID: PMC11956161 DOI: 10.3389/fonc.2025.1521204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/21/2025] [Indexed: 04/02/2025] Open
Abstract
Background Prostate cancer is defined by the suppression of genes that suppress tumours and the activation of proto-oncogenes. These are the hallmarks of prostate cancer, and they have been linked to numerous genomic variations, which lead to unfavourable treatment outcomes. Prostate cancer can be categorised into various risk groups of tumour molecular subtypes grounded in the idea of genomic structural variations connected to TMPRSS2:ERG fusion and loss of PTEN. Research suggests that certain genomic alterations may be more prevalent or exhibit different patterns in prostate cancer tumours across populations. Studies have reported a higher frequency of PTEN loss and TMPRSS2:ERG fusion in prostate tumours of Black/African American men, which may contribute to the more aggressive nature of the disease in this population. Thus, therapeutically important information can be obtained from these structural variations, including correlations with poor prognosis and disease severity. Methods Peer-reviewed articles from 1998 to 2024 were sourced from PubMed and Google Scholar. During the review process, the following search terms were employed: "Tumour suppressor genes OR variations OR alterations OR oncogenes OR diagnostics OR ethnicity OR biomarkers OR prostate cancer genomics OR prostate cancer structural variations OR tumour and molecular subtypes OR therapeutic implications OR immunotherapy OR immunogenetics." Results There was a total of 13,012 results for our search query: 5,903 publications from Google Scholar with the patent and citation unchecked filer options, and 7127 articles from PubMed with the abstract, free full text, and full-text options selected. Unpublished works were not involved. Except for four articles published between 1998 and 1999, all other selected articles published in 2000 and later were considered. However, papers with irrelevant information or redundant or duplicate content were not chosen for this review. Thus, 134 met the inclusion criteria and were ultimately retained for this review. Conclusion This review extracted 134 relevant articles about genomic structure variations in prostate cancer. Our findings demonstrate the importance of PTEN and TMPRSS2:ERG fusion and tumour molecular subtyping in prostate cancer precision medicine.
Collapse
Affiliation(s)
- Fidelis Charles Bugoye
- Directorate of Forensic Science and DNA Services, Government Chemist Laboratory Authority, Dar es Salaam, Tanzania
- Department of Pathology, Moi University, Moi Teaching and Referral Hospital, Eldoret, Kenya
| | - Rispah Torrorey-Sawe
- Department of Pathology, Moi University, Moi Teaching and Referral Hospital, Eldoret, Kenya
| | - Richard Biegon
- Department of Pathology, Moi University, Moi Teaching and Referral Hospital, Eldoret, Kenya
| | - Nazima Dharsee
- Clinical Research, Training and Consultancy Unit, Ocean Road Cancer Institute, Dar es Salaam, Tanzania
| | - Fidelice Mafumiko
- Directorate of Forensic Science and DNA Services, Government Chemist Laboratory Authority, Dar es Salaam, Tanzania
| | - Herry Kibona
- Department of Urology, Muhimbili National Hospital, Dar es Salaam, Tanzania
| | - Said Aboud
- Head Office, National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Kirtika Patel
- Department of Pathology, Moi University, Moi Teaching and Referral Hospital, Eldoret, Kenya
| | - Simeon Mining
- Department of Pathology, Moi University, Moi Teaching and Referral Hospital, Eldoret, Kenya
| |
Collapse
|
3
|
Pedrani M, Barizzi J, Salfi G, Nepote A, Testi I, Merler S, Castelo-Branco L, Mestre RP, Turco F, Tortola L, Theurillat JP, Gillessen S, Vogl U. The Emerging Predictive and Prognostic Role of Aggressive-Variant-Associated Tumor Suppressor Genes Across Prostate Cancer Stages. Int J Mol Sci 2025; 26:318. [PMID: 39796175 PMCID: PMC11719667 DOI: 10.3390/ijms26010318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/23/2024] [Accepted: 12/31/2024] [Indexed: 01/13/2025] Open
Abstract
Aggressive variant prostate cancer (AVPC) is characterized by a molecular signature involving combined defects in TP53, RB1, and/or PTEN (AVPC-TSGs), identifiable through immunohistochemistry or genomic analysis. The reported prevalence of AVPC-TSG alterations varies widely, reflecting differences in assay sensitivity, treatment pressure, and disease stage evolution. Although robust clinical evidence is still emerging, the study of AVPC-TSG alterations in prostate cancer (PCa) is promising. Alterations in TP53, RB1, and PTEN, as well as the combined loss of AVPC-TSGs, may have significant implications for prognosis and treatment. These biomarkers might help predict responses to various therapies, including hormonal treatments, cytotoxic agents, radiotherapy, and targeted therapies. Understanding the impact of these molecular alterations in patients with PCa is crucial for personalized management. In this review, we provide a comprehensive overview of the emerging prognostic and predictive roles of AVPC-TSG alterations across PCa stages. Moreover, we discuss the implications of different methods used for detecting AVPC-TSG alterations and summarize factors influencing their prevalence. As our comprehension of the genomic landscape of PCa disease deepens, incorporating genomic profiling into clinical decision making will become increasingly important for improving patient outcomes.
Collapse
Affiliation(s)
- Martino Pedrani
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.P.); (A.N.); (S.M.); (R.P.M.); (F.T.); (S.G.)
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, 20122 Milan, Italy
| | - Jessica Barizzi
- Istituto Cantonale di Patologia, Ente Ospedaliero Cantonale (EOC), 6600 Locarno, Switzerland
| | - Giuseppe Salfi
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.P.); (A.N.); (S.M.); (R.P.M.); (F.T.); (S.G.)
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland;
| | - Alessandro Nepote
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.P.); (A.N.); (S.M.); (R.P.M.); (F.T.); (S.G.)
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland;
- AOU San Luigi Gonzaga, Department of Oncology, University of Torino, 10124 Torino, Italy
| | - Irene Testi
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.P.); (A.N.); (S.M.); (R.P.M.); (F.T.); (S.G.)
- Medical Oncology Unit, University Hospital of Parma, 43126 Parma, Italy
| | - Sara Merler
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.P.); (A.N.); (S.M.); (R.P.M.); (F.T.); (S.G.)
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland;
- Section of Innovation Biomedicine—Oncology Area, Department of Engineering for Innovation Medicine, University of Verona and Verona University Hospital Trust, 37126 Verona, Italy
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| | - Luis Castelo-Branco
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.P.); (A.N.); (S.M.); (R.P.M.); (F.T.); (S.G.)
| | - Ricardo Pereira Mestre
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.P.); (A.N.); (S.M.); (R.P.M.); (F.T.); (S.G.)
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland;
| | - Fabio Turco
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.P.); (A.N.); (S.M.); (R.P.M.); (F.T.); (S.G.)
| | - Luigi Tortola
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.P.); (A.N.); (S.M.); (R.P.M.); (F.T.); (S.G.)
| | - Jean-Philippe Theurillat
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland;
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| | - Silke Gillessen
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.P.); (A.N.); (S.M.); (R.P.M.); (F.T.); (S.G.)
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| | - Ursula Vogl
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.P.); (A.N.); (S.M.); (R.P.M.); (F.T.); (S.G.)
| |
Collapse
|
4
|
Yu F, Zeng G, Yang L, Zhou H, Wang Y. LAMB3: Central role and clinical significance in neoplastic and non-neoplastic diseases. Biomed Pharmacother 2024; 178:117233. [PMID: 39111076 DOI: 10.1016/j.biopha.2024.117233] [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: 05/25/2024] [Revised: 07/18/2024] [Accepted: 07/30/2024] [Indexed: 08/25/2024] Open
Abstract
Recently, topics related to targeted gene therapy and diagnosis have become increasingly important in disease research. The progression of many diseases is associated with specific gene signaling pathways. Therefore, the identification of precise gene targets in various diseases is crucial for the development of effective treatments. Laminin subunit beta 3 (LAMB3), a component of laminin 5, functions as an adhesive protein in the extracellular matrix and plays a vital role in regulating cell proliferation, migration, and cell cycle in certain diseases. Previous studies have indicated that LAMB3 is highly expressed in numerous tumorous and non-tumorous conditions, including renal fibrosis; squamous cell carcinoma of the skin, thyroid, lung, pancreatic, ovarian, colorectalr, gastric, breast, cervical, nasopharyngeal, bladder, prostate cancers; and cholangiocarcinoma. Conversely, it is underexpressed in other conditions, such as hepatocellular carcinoma, epidermolysis bullosa, and amelogenesis imperfecta. Consequently, LAMB3 may serve as a molecular diagnostic and therapeutic target for various diseases through its involvement in critical gene signaling pathways. This paper reviews the research status of LAMB3 and its role in related diseases.
Collapse
Affiliation(s)
- Fangqiu Yu
- Urological Department, First Hospital of Jilin University, Changchun, Jilin Province 130021, China
| | - Guoqiang Zeng
- Urological Department, First Hospital of Jilin University, Changchun, Jilin Province 130021, China
| | - Lei Yang
- Urological Department, First Hospital of Jilin University, Changchun, Jilin Province 130021, China
| | - Honglan Zhou
- Urological Department, First Hospital of Jilin University, Changchun, Jilin Province 130021, China
| | - Yuantao Wang
- Urological Department, First Hospital of Jilin University, Changchun, Jilin Province 130021, China.
| |
Collapse
|
5
|
Smith TAD. Gene Abnormalities and Modulated Gene Expression Associated with Radionuclide Treatment: Towards Predictive Biomarkers of Response. Genes (Basel) 2024; 15:688. [PMID: 38927624 PMCID: PMC11202453 DOI: 10.3390/genes15060688] [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: 05/01/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
Molecular radiotherapy (MRT), also known as radioimmunotherapy or targeted radiotherapy, is the delivery of radionuclides to tumours by targeting receptors overexpressed on the cancer cell. Currently it is used in the treatment of a few cancer types including lymphoma, neuroendocrine, and prostate cancer. Recently reported outcomes demonstrating improvements in patient survival have led to an upsurge in interest in MRT particularly for the treatment of prostate cancer. Unfortunately, between 30% and 40% of patients do not respond. Further normal tissue exposure, especially kidney and salivary gland due to receptor expression, result in toxicity, including dry mouth. Predictive biomarkers to select patients who will benefit from MRT are crucial. Whilst pre-treatment imaging with imaging versions of the therapeutic agents is useful in demonstrating tumour binding and potentially organ toxicity, they do not necessarily predict patient benefit, which is dependent on tumour radiosensitivity. Transcript-based biomarkers have proven useful in tailoring external beam radiotherapy and adjuvant treatment. However, few studies have attempted to derive signatures for MRT response prediction. Here, transcriptomic studies that have identified genes associated with clinical radionuclide exposure have been reviewed. These studies will provide potential features for seeding multi-component biomarkers of MRT response.
Collapse
Affiliation(s)
- Tim A D Smith
- Nuclear Futures Institute, School of Computer Science and Engineering, Bangor University, Dean Street, Bangor LL57 1UT, UK
| |
Collapse
|
6
|
Eberlein C, Williamson SC, Hopcroft L, Ros S, Moss JI, Kerr J, van Weerden WM, de Bruin EC, Dunn S, Willis B, Ross SJ, Rooney C, Barry ST. Capivasertib combines with docetaxel to enhance anti-tumour activity through inhibition of AKT-mediated survival mechanisms in prostate cancer. Br J Cancer 2024; 130:1377-1387. [PMID: 38396173 PMCID: PMC11014923 DOI: 10.1038/s41416-024-02614-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/25/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND/OBJECTIVE To explore the anti-tumour activity of combining AKT inhibition and docetaxel in PTEN protein null and WT prostate tumours. METHODS Mechanisms associated with docetaxel capivasertib treatment activity in prostate cancer were examined using a panel of in vivo tumour models and cell lines. RESULTS Combining docetaxel and capivasertib had increased activity in PTEN null and WT prostate tumour models in vivo. In vitro short-term docetaxel treatment caused cell cycle arrest in the majority of cells. However, a sub-population of docetaxel-persister cells did not undergo G2/M arrest but upregulated phosphorylation of PI3K/AKT pathway effectors GSK3β, p70S6K, 4E-BP1, but to a lesser extent AKT. In vivo acute docetaxel treatment induced p70S6K and 4E-BP1 phosphorylation. Treating PTEN null and WT docetaxel-persister cells with capivasertib reduced PI3K/AKT pathway activation and cell cycle progression. In vitro and in vivo it reduced proliferation and increased apoptosis or DNA damage though effects were more marked in PTEN null cells. Docetaxel-persister cells were partly reliant on GSK3β as a GSK3β inhibitor AZD2858 reversed capivasertib-induced apoptosis and DNA damage. CONCLUSION Capivasertib can enhance anti-tumour effects of docetaxel by targeting residual docetaxel-persister cells, independent of PTEN status, to induce apoptosis and DNA damage in part through GSK3β.
Collapse
Affiliation(s)
- Cath Eberlein
- Bioscience, Early Oncology, AstraZeneca, Alderley Park, UK
| | | | | | - Susana Ros
- Bioscience, Early Oncology, AstraZeneca, Cambridge, UK
| | | | - James Kerr
- Bioscience, Early Oncology, AstraZeneca, Cambridge, UK
| | - Wytske M van Weerden
- Department of Experimental Urology, Josephine Nefkens Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - Shanade Dunn
- Bioscience, Early Oncology, AstraZeneca, Cambridge, UK
| | - Brandon Willis
- Bioscience, Early Oncology, AstraZeneca, Boston, MA, USA
| | - Sarah J Ross
- Bioscience, Early Oncology, AstraZeneca, Cambridge, UK
| | | | - Simon T Barry
- Bioscience, Early Oncology, AstraZeneca, Cambridge, UK.
| |
Collapse
|
7
|
Gupta S, To TM, Graf R, Kadel EE, Reilly N, Albarmawi H. Real-World Overall Survival and Treatment Patterns by PTEN Status in Metastatic Castration-Resistant Prostate Cancer. JCO Precis Oncol 2024; 8:e2300562. [PMID: 38547419 PMCID: PMC10994466 DOI: 10.1200/po.23.00562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/16/2024] [Accepted: 02/07/2024] [Indexed: 04/02/2024] Open
Abstract
PURPOSE It is estimated that the PTEN tumor suppressor gene is functionally lost in 40%-50% of patients with metastatic castration-resistant prostate cancer (mCRPC). There is limited information on the prognostic significance of PTEN status identified with genomic testing. This real-world cohort study assessed PTEN as a genetic biomarker using data from US-based oncology practices. METHODS This retrospective real-world cohort study used a deidentified US-based metastatic prostate cancer clinicogenomic database linked to longitudinal clinical data derived from electronic health records. Patients were aged 18 years and older and diagnosed with mCRPC between January 1, 2018, and June 30, 2021. Comprehensive genomic profiling (CGP) of tumor specimens was performed using next-generation sequencing. First-line (1L) and second-line (2L) treatment patterns were assessed and stratified by PTEN status. Kaplan-Meier methods and a multivariable Cox model were used to compare the real-world overall survival by PTEN status among patients who received 1L novel hormone therapy or taxanes. RESULTS In patients with mCRPC who underwent CGP, PTEN loss of function (LOF) was associated with decreased survival compared with intact PTEN (hazard ratio, 1.61 [95% CI, 1.07 to 2.42]; P = .024). The results were not influenced by 1L treatment type. 1L treatment patterns were similar between intact PTEN and PTEN LOF subgroups, with abiraterone and enzalutamide being the two most common treatments in both groups. Patients with PTEN LOF were less likely to receive 2L treatments than patients with intact PTEN. CONCLUSION PTEN LOF, identified with genomic testing, was associated with decreased survival and negative prognoses in patients with mCRPC.
Collapse
Affiliation(s)
- Shilpa Gupta
- Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH
| | - Tu My To
- Genentech, Inc, South San Francisco, CA
| | - Ryon Graf
- Foundation Medicine, Inc, Cambridge, MA
| | | | | | | |
Collapse
|
8
|
Maekawa S, Takata R, Obara W. Molecular Mechanisms of Prostate Cancer Development in the Precision Medicine Era: A Comprehensive Review. Cancers (Basel) 2024; 16:523. [PMID: 38339274 PMCID: PMC10854717 DOI: 10.3390/cancers16030523] [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: 11/24/2023] [Revised: 01/21/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
The progression of prostate cancer (PCa) relies on the activation of the androgen receptor (AR) by androgens. Despite efforts to block this pathway through androgen deprivation therapy, resistance can occur through several mechanisms, including the abnormal activation of AR, resulting in castration-resistant PCa following the introduction of treatment. Mutations, amplifications, and splicing variants in AR-related genes have garnered attention in this regard. Furthermore, recent large-scale next-generation sequencing analysis has revealed the critical roles of AR and AR-related genes, as well as the DNA repair, PI3K, and cell cycle pathways, in the onset and progression of PCa. Moreover, research on epigenomics and microRNA has increasingly become popular; however, it has not translated into the development of effective therapeutic strategies. Additionally, treatments targeting homologous recombination repair mutations and the PI3K/Akt pathway have been developed and are increasingly accessible, and multiple clinical trials have investigated the efficacy of immune checkpoint inhibitors. In this comprehensive review, we outline the status of PCa research in genomics and briefly explore potential future developments in the field of epigenetic modifications and microRNAs.
Collapse
Affiliation(s)
- Shigekatsu Maekawa
- Department of Urology, Iwate Medical University, Iwate 028-3694, Japan; (R.T.); (W.O.)
| | | | | |
Collapse
|
9
|
Erak E, Oliveira LD, Mendes AA, Dairo O, Ertunc O, Kulac I, Baena-Del Valle JA, Jones T, Hicks JL, Glavaris S, Guner G, Vidal ID, Markowski M, de la Calle C, Trock BJ, Meena A, Joshi U, Kondragunta C, Bonthu S, Singhal N, De Marzo AM, Lotan TL. Predicting Prostate Cancer Molecular Subtype With Deep Learning on Histopathologic Images. Mod Pathol 2023; 36:100247. [PMID: 37307876 PMCID: PMC11225718 DOI: 10.1016/j.modpat.2023.100247] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 06/06/2023] [Indexed: 06/14/2023]
Abstract
Microscopic examination of prostate cancer has failed to reveal a reproducible association between molecular and morphologic features. However, deep-learning algorithms trained on hematoxylin and eosin (H&E)-stained whole slide images (WSI) may outperform the human eye and help to screen for clinically-relevant genomic alterations. We created deep-learning algorithms to identify prostate tumors with underlying ETS-related gene (ERG) fusions or PTEN deletions using the following 4 stages: (1) automated tumor identification, (2) feature representation learning, (3) classification, and (4) explainability map generation. A novel transformer-based hierarchical architecture was trained on a single representative WSI of the dominant tumor nodule from a radical prostatectomy (RP) cohort with known ERG/PTEN status (n = 224 and n = 205, respectively). Two distinct vision transformer-based networks were used for feature extraction, and a distinct transformer-based model was used for classification. The ERG algorithm performance was validated across 3 RP cohorts, including 64 WSI from the pretraining cohort (AUC, 0.91) and 248 and 375 WSI from 2 independent RP cohorts (AUC, 0.86 and 0.89, respectively). In addition, we tested the ERG algorithm performance in 2 needle biopsy cohorts comprised of 179 and 148 WSI (AUC, 0.78 and 0.80, respectively). Focusing on cases with homogeneous (clonal) PTEN status, PTEN algorithm performance was assessed using 50 WSI reserved from the pretraining cohort (AUC, 0.81), 201 and 337 WSI from 2 independent RP cohorts (AUC, 0.72 and 0.80, respectively), and 151 WSI from a needle biopsy cohort (AUC, 0.75). For explainability, the PTEN algorithm was also applied to 19 WSI with heterogeneous (subclonal) PTEN loss, where the percentage tumor area with predicted PTEN loss correlated with that based on immunohistochemistry (r = 0.58, P = .0097). These deep-learning algorithms to predict ERG/PTEN status prove that H&E images can be used to screen for underlying genomic alterations in prostate cancer.
Collapse
Affiliation(s)
- Eric Erak
- Department of Pathology, Johns Hopkins University School of Medicine
| | | | - Adrianna A Mendes
- Department of Pathology, Johns Hopkins University School of Medicine
| | | | - Onur Ertunc
- Department of Pathology, Suleyman Demirel University, Turkey
| | | | | | - Tracy Jones
- Department of Pathology, Johns Hopkins University School of Medicine
| | - Jessica L Hicks
- Department of Pathology, Johns Hopkins University School of Medicine
| | | | | | | | - Mark Markowski
- Department of Oncology, Johns Hopkins University School of Medicine
| | | | - Bruce J Trock
- Department of Urology, Johns Hopkins University School of Medicine
| | | | | | | | | | | | - Angelo M De Marzo
- Department of Pathology, Johns Hopkins University School of Medicine; Department of Oncology, Johns Hopkins University School of Medicine; Department of Urology, Johns Hopkins University School of Medicine
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins University School of Medicine; Department of Oncology, Johns Hopkins University School of Medicine; Department of Urology, Johns Hopkins University School of Medicine.
| |
Collapse
|
10
|
Yang J, Xin C, Yin G, Li J. Taraxasterol suppresses the proliferation and tumor growth of androgen-independent prostate cancer cells through the FGFR2-PI3K/AKT signaling pathway. Sci Rep 2023; 13:13072. [PMID: 37567936 PMCID: PMC10421874 DOI: 10.1038/s41598-023-40344-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 08/09/2023] [Indexed: 08/13/2023] Open
Abstract
Prostate cancer (PCa) is prevalent among older men and difficult to survive after metastasis. It is urgent to find new drugs and treatments. Several studies show that taraxasterol (TAX) has important anti-inflammatory, anti-oxidative and anti-tumor effects. However, the function and mechanisms of TAX in PCa remain unclear. Here, we found that TAX could significantly suppress the viability and growth of androgen-independent PCa cells and down-regulate the expression of c-Myc and cyclin D1 in vitro. Mechanistically, PI3K/AKT signaling pathway was weakened and the expression of FGFR2 was reduced after TAX treatment in androgen-independent PCa cells. Moreover, TAX evidently inhibited the tumor growth in nude mice and the expression of c-Myc, cyclin D1, p-AKT and FGFR2 were down-regulated in xenograft tumor. These results indicate that TAX suppresses the proliferation of androgen-independent PCa cells via inhibiting the activation of PI3K/AKT signaling pathway and the expression of FGFR2, which means TAX may be a novel anti-tumor agent for later PCa treatment.
Collapse
Affiliation(s)
- Jinqiu Yang
- School of Clinical Medicine, Dali University, Dali, 671013, Yunnan, China
| | - Chulin Xin
- School of Basic Medical Sciences, Dali University, 22 Wanhua Road, Dali, 671013, Yunnan, China
| | - Guangfen Yin
- The First Affiliated Hospital of Dali University, Dali, 671013, Yunnan, China
| | - Juan Li
- School of Basic Medical Sciences, Dali University, 22 Wanhua Road, Dali, 671013, Yunnan, China.
| |
Collapse
|
11
|
Salles DC, Mendes AA, Han M, Partin AW, Trock BJ, Jing Y, Lotan TL. ERG Status at the Margin Is Associated With Biochemical Recurrence After Radical Prostatectomy With Positive Surgical Margins. Mod Pathol 2023; 36:100147. [PMID: 36828362 PMCID: PMC10442458 DOI: 10.1016/j.modpat.2023.100147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/22/2022] [Accepted: 02/16/2023] [Indexed: 02/24/2023]
Abstract
Positive surgical margins at radical prostatectomy are associated with an increased risk of biochemical recurrence (BCR). However, there is considerable variability in outcomes, suggesting that molecular biomarkers-when assessed specifically at the margin tumor tissue-may be useful to stratify prognosis in this group. We used a case-cohort design for the outcome of BCR, selecting 215 patients from a cohort of 813 patients undergoing prostatectomy treated at the Johns Hopkins from 2008 to 2017 with positive margins and available clinical data. Tissue microarrays were created from the tumor adjacent to the positive margin and stained for PTEN, ERG, and Ki-67. Cases were scored dichotomously (PTEN and ERG) or by the Ki-67 staining index using previously validated protocols. The analysis used Cox proportional hazards models weighted for the case-cohort design. Overall, 20% (37/185) of evaluable cases had PTEN loss and 38% (71/185) had ERG expression, and the median Ki-67 expression was 0.42%. In multivariable analysis adjusting for the CAPRA-S score, adjuvant radiation, and grade group at the positive margin, ERG-positive tumors were associated with a higher risk of BCR compared to those that were ERGnegative (hazard ratio [HR], 2.4; 95% CI, 1.2-4.9; P = .012) regardless of PTEN status at the margin, and adding ERG to clinicopathologic variables increased the concordance index from 0.827 to 0.847. PTEN loss was associated with an increased risk of BCR on univariable analysis (HR, 3.19; 95% CI, 1.72-5.92; P = .0002), but this association did not remain after adjusting for clinicopathologic variables (HR, 1.06; 95% CI, 0.49-2.29; P = .890). Thus, in the setting of prostate tumors with positive surgical margins after prostatectomy, ERG-positive tumors with or without PTEN loss at the positive margin are associated with a significantly higher risk of BCR after adjusting for clinicopathologic variables. If validated, ERG status may be helpful in decision-making surrounding adjuvant therapy after prostatectomy.
Collapse
Affiliation(s)
- Daniela C Salles
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Adrianna A Mendes
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Misop Han
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alan W Partin
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Bruce J Trock
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yuezhou Jing
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
12
|
Voulgari O, Goutas D, Pergaris A, Belogiannis K, Thymara E, Kavantzas N, Lazaris AC. Correlations of PTEN and ERG Immunoexpression in Prostate Carcinoma and Lesions Related to Its Natural History: Clinical Perspectives. Curr Issues Mol Biol 2023; 45:2767-2780. [PMID: 37185705 PMCID: PMC10136580 DOI: 10.3390/cimb45040181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Purpose: The aim of our study was to observe the associations between the ETS-related gene (ERG) and the phosphatase and tensin homolog gene (PTEN) immunoexpression in prostate cancer and related lesions and highlight the clinical significance of these findings. Methods: We evaluated the immunohistochemical expression of ERG and PTEN in a series of 151 invasive prostate adenocarcinomas, including low-grade (Gleason grade pattern 3) and high-grade (Gleason grade patterns 4, 5) morphological patterns which corresponded to 45.5% and 54.4% of the cases, respectively. Additionally, we evaluated the immunoexpression of the two markers both in foci of high-grade prostatic intraepithelial neoplasia (HGPIN), as a precursor lesion of cancer, and in foci of intraductal carcinoma of the prostate (IDCP). Finally, to ensure the malignant nature of the prostate glands examined, we employed p63 and alpha-methylacyl-CoA racemase (AMACR) expression. Results: We found that PTEN loss was observed in 50.7%, and ERG positivity was detected in 41.8% of our cancerous samples. In HGPIN, PTEN loss appeared to be linked with a high-grade adjacent invasive carcinoma component which also displayed PTEN loss. As far as IDCP is concerned, ERG immunonegativity was correlated with adjacent high-grade invasive cancer, which was also ERG immunonegative. Conclusions: Our findings suggest that the clonal expansion of invasive cancer appears to be associated with distinct immunophenotypic cellular alterations of both early and late cancer-related histological lesions. Patients with PTEN loss in HGPIN in prostate biopsies should be closely monitored due to the increased likelihood of having an associated invasive high-grade carcinoma that may have not been sampled. Given the clinical significance that derives from PTEN expression in HGPIN lesions, we suggest the routine use of PTEN immunohistochemistry in prostate cancer biopsies in which HGPIN is the only finding.
Collapse
|
13
|
Characterization of exposure–response relationships of ipatasertib in patients with metastatic castration-resistant prostate cancer in the IPATential150 study. Cancer Chemother Pharmacol 2022; 90:511-521. [PMID: 36305957 PMCID: PMC9637074 DOI: 10.1007/s00280-022-04488-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/19/2022] [Indexed: 12/02/2022]
Abstract
Purpose The exposure–response relationships for efficacy and safety of ipatasertib, a selective AKT kinase inhibitor, were characterized using data collected from 1101 patients with metastatic castration-resistant prostate cancer in the IPATential150 study (NCT03072238). Methods External validation of a previously developed population pharmacokinetic model was performed using the observed pharmacokinetic data from the IPATential150 study. Exposure metrics of ipatasertib for subjects who received ipatasertib 400 mg once-daily orally in this study were generated as model-predicted area under the concentration–time curve at steady state (AUCSS). The exposure–response relationship with radiographic progression-free survival (rPFS) was evaluated using Cox regression and relationships with safety endpoints were assessed using logistic regression. Results A statistically significant correlation between ipatasertib AUCSS and improved survival was found in patients with PTEN-loss tumors (hazard ratio [HR]: 0.92 per 1000 ng h/mL AUCSS, 95% confidence interval [CI] 0.87–0.98, p = 0.011). In contrast, an improvement in rPFS was seen in subjects receiving ipatasertib treatment (HR: 0.84, 95% CI 0.71–0.99, p = 0.038) but this effect was not associated with ipatasertib AUCSS in the intention-to-treat population. Incidences of some adverse events (AEs) had statistically significant association with ipatasertib AUCSS (serious AEs, AEs leading to discontinuation, and Grade ≥ 2 hyperglycemia), while others were associated with only ipatasertib treatment (AEs leading to dose reduction, Grade ≥ 3 diarrhea, and Grade ≥ 2 rash). Conclusions The exposure–efficacy results indicated that patients receiving ipatasertib may continue benefiting from this treatment at the administered dose, despite some variability in exposures, while the exposure–safety results suggested increased risks of AEs with ipatasertib treatment and/or increased ipatasertib exposures. Supplementary Information The online version contains supplementary material available at 10.1007/s00280-022-04488-2.
Collapse
|
14
|
Wise HM, Harris A, Kriplani N, Schofield A, Caldwell H, Arends MJ, Overton IM, Leslie NR. PTEN Protein Phosphatase Activity Is Not Required for Tumour Suppression in the Mouse Prostate. Biomolecules 2022; 12:1511. [PMID: 36291720 PMCID: PMC9599176 DOI: 10.3390/biom12101511] [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: 09/22/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 12/02/2022] Open
Abstract
Loss PTEN function is one of the most common events driving aggressive prostate cancers and biochemically, PTEN is a lipid phosphatase which opposes the activation of the oncogenic PI3K-AKT signalling network. However, PTEN also has additional potential mechanisms of action, including protein phosphatase activity. Using a mutant enzyme, PTEN Y138L, which selectively lacks protein phosphatase activity, we characterised genetically modified mice lacking either the full function of PTEN in the prostate gland or only lacking protein phosphatase activity. The phenotypes of mice carrying a single allele of either wild-type Pten or PtenY138L in the prostate were similar, with common prostatic intraepithelial neoplasia (PIN) and similar gene expression profiles. However, the latter group, lacking PTEN protein phosphatase activity additionally showed lymphocyte infiltration around PIN and an increased immune cell gene expression signature. Prostate adenocarcinoma, elevated proliferation and AKT activation were only frequently observed when PTEN was fully deleted. We also identify a common gene expression signature of PTEN loss conserved in other studies (including Nkx3.1, Tnf and Cd44). We provide further insight into tumour development in the prostate driven by loss of PTEN function and show that PTEN protein phosphatase activity is not required for tumour suppression.
Collapse
Affiliation(s)
- Helen M. Wise
- Institute of Biological Chemistry, Biophysics and Bioengineering, Riccarton Campus, Heriot Watt University, Nasmyth Building, Edinburgh EH14 4AS, UK
| | - Adam Harris
- Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University, 97 Lisburn Road, Belfast BT9 7AE, UK
| | - Nisha Kriplani
- Institute of Biological Chemistry, Biophysics and Bioengineering, Riccarton Campus, Heriot Watt University, Nasmyth Building, Edinburgh EH14 4AS, UK
| | - Adam Schofield
- Institute of Biological Chemistry, Biophysics and Bioengineering, Riccarton Campus, Heriot Watt University, Nasmyth Building, Edinburgh EH14 4AS, UK
| | - Helen Caldwell
- Edinburgh Pathology, Cancer Research UK Edinburgh Centre, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh EH4 2XR, UK
| | - Mark J. Arends
- Edinburgh Pathology, Cancer Research UK Edinburgh Centre, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh EH4 2XR, UK
| | - Ian M. Overton
- Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University, 97 Lisburn Road, Belfast BT9 7AE, UK
| | - Nick R. Leslie
- Institute of Biological Chemistry, Biophysics and Bioengineering, Riccarton Campus, Heriot Watt University, Nasmyth Building, Edinburgh EH14 4AS, UK
| |
Collapse
|
15
|
Stephen N, Badhe BA. Diagnostic utility of immunohistochemical markers alpha methyl acyl coA racemase (AMACR) and Ets related gene (ERG) in prostate cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2022; 15:364-372. [PMID: 36237639 PMCID: PMC9547992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 06/21/2022] [Indexed: 06/16/2023]
Abstract
OBJECTIVES To study the sensitivity and specificity of IHC markers AMACR and ERG in prostatic adenocarcinoma. METHODS The study was a prospective one and samples were collected from August 2014 to June 2016. A total of 186 samples were obtained from the Department of Urology, in which 112 of these were benign prostatic hyperplasia (BPH), and 71 were prostatic adenocarcinoma. The adenocarcinoma cases were evaluated by two histopathologists, and appropriate Gleason score was given according to the modified ISUP Gleason grading system (2016). IHC markers AMACR & ERG were performed on the adenocarcinoma cases and their sensitivity and specificity were calculated. RESULTS AMACR was a highly sensitive and specific marker for detecting prostatic carcinoma with a sensitivity and specificity of 95.8% and 96.5% respectively. ERG was a very specific marker with poor sensitivity in detecting prostate cancer. The sensitivity and specificity of ERG were 35.2% and 100% respectively. ERG expression decreased with increasing Gleason grade, PSA level, and tumour volume, which was statistically significant while the association of AMACR with Gleason grade or with tumor volume was not significant. CONCLUSION ERG is a marker of early prostatic carcinogenesis and tumors may be positive or negative subtypes. Special histomorphologic features like perineural invasion, glomerulations, and intraluminal blue mucin were also studied. AMACR was a highly sensitive marker for detecting prostatic adenocarcinoma, while ERG was highly specific.
Collapse
Affiliation(s)
- Norton Stephen
- Department of Pathology, Dhanvantri Nagar, Gorimedu, Puducherry 605 006, JIPMER India
| | - Bhawana A Badhe
- Department of Pathology, Dhanvantri Nagar, Gorimedu, Puducherry 605 006, JIPMER India
| |
Collapse
|
16
|
Solamargine Inhibits Prostate Cancer Cell Growth and Enhances the Therapeutic Efficacy of Docetaxel via Akt Signaling. JOURNAL OF ONCOLOGY 2022; 2022:9055954. [PMID: 35310915 PMCID: PMC8930254 DOI: 10.1155/2022/9055954] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 01/20/2022] [Accepted: 02/16/2022] [Indexed: 12/12/2022]
Abstract
Prostate cancer (PCa) has become a leading cause of cancer-associated incidence and mortality in men worldwide. However, most primary PCas relapse to castration-resistant PCa (CRPC) after androgen deprivation treatment. The current treatment for CRPC is based on chemotherapeutic drugs such as docetaxel, while the development of chemoresistance and severe side effects limit the therapeutic benefit. Solamargine, a natural alkaloid isolated from a traditional Chinese herbal medicine known as Solanum nigrum, exhibits antitumor activity in various human cancers. In this study, we demonstrated that solamargine substantially inhibited CRPC cell growth in a dose-dependent manner through the suppression of phosphoinositide 3-kinase (PI3K)/Akt signaling. Moreover, solamargine exhibited significant antitumor effects in mouse xenograft models. Bioinformatics analysis of docetaxel-resistant PCa cells indicated that the PI3K/Akt pathway mediated the chemoresistance of CRPC. Furthermore, solamargine significantly enhanced the efficacy of docetaxel in PCa cells. These results reveal the therapeutic potential of solamargine against human PCa.
Collapse
|
17
|
Gasmi A, Roubaud G, Dariane C, Barret E, Beauval JB, Brureau L, Créhange G, Fiard G, Fromont G, Gauthé M, Ruffion A, Renard-Penna R, Sargos P, Rouprêt M, Ploussard G, Mathieu R. Overview of the Development and Use of Akt Inhibitors in Prostate Cancer. J Clin Med 2021; 11:jcm11010160. [PMID: 35011901 PMCID: PMC8745410 DOI: 10.3390/jcm11010160] [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: 12/04/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 12/15/2022] Open
Abstract
Deregulation of the PI3K-Akt-mTOR pathway plays a critical role in the development and progression of many cancers. In prostate cancer, evidence suggests that it is mainly driven by PTEN loss of function. For many years, the development of selective Akt inhibitors has been challenging. In recent phase II and III clinical trials, Ipatasertib and Capivasertib associated with androgen deprivation therapies showed promising outcomes in patients with metastatic castration-resistant prostate cancer and PTEN-loss. Ongoing trials are currently assessing several Akt inhibitors in prostate cancer with different combinations, at different stages of the disease.
Collapse
Affiliation(s)
- Anis Gasmi
- Department of Urology, University of Rennes, 35000 Rennes, France;
- Correspondence:
| | - Guilhem Roubaud
- Department of Medical Oncology, Institut Bergonié, 33000 Bordeaux, France;
| | - Charles Dariane
- Department of Urology, Hôpital Européen Georges-Pompidou, AP-HP, Paris University, 75005 Paris, France;
| | - Eric Barret
- Department of Urology, Institut Mutualiste Montsouris, 75005 Paris, France;
| | - Jean-Baptiste Beauval
- Department of Urology, La Croix du Sud Hôpital, Quint Fonsegrives, 31000 Toulouse, France; (J.-B.B.); (G.P.)
| | - Laurent Brureau
- Department of Urology, CHU de Pointe-à-Pitre, University of Antilles, 97110 Pointe-à-Pitre, France;
| | - Gilles Créhange
- Department of Urology, University Hospital, Université Grenoble Alpes, 38000 Grenoble, France;
| | - Gaëlle Fiard
- Department of Radiation Oncology, Curie Institute, 75005 Paris, France;
| | - Gaëlle Fromont
- Department of Pathology, CHRU Tours, 37000 Tours, France;
| | - Mathieu Gauthé
- Department of Nuclear Medicine, Scintep, 38000 Grenoble, France;
| | - Alain Ruffion
- Service d’Urologie Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, 69000 Lyon, France;
- Equipe 2, Centre d’Innovation en Cancérologie de Lyon (EA 3738 CICLY), Faculté de Médecine Lyon Sud, Université Lyon 1, 69000 Lyon, France
| | - Raphaële Renard-Penna
- Department of Radiology, Sorbonne University, AP-HP, Pitie-Salpetriere Hospital, 75013 Paris, France;
| | - Paul Sargos
- Department of Radiotherapy, Institut Bergonié, 33000 Bordeaux, France;
| | - Morgan Rouprêt
- Department of Urology, Sorbonne University, GRC 5 Predictive Onco-Uro, AP-HP, Urology, Pitie-Salpetriere Hospital, 75013 Paris, France;
| | - Guillaume Ploussard
- Department of Urology, La Croix du Sud Hôpital, Quint Fonsegrives, 31000 Toulouse, France; (J.-B.B.); (G.P.)
| | - Romain Mathieu
- Department of Urology, University of Rennes, 35000 Rennes, France;
- IRSET (Institut de Recherche en Santé, Environnement et Travail), University of Rennes, Inserm, EHESP, 35000 Rennes, France
| |
Collapse
|
18
|
Mollica V, Marchetti A, Rosellini M, Nuvola G, Rizzo A, Santoni M, Cimadamore A, Montironi R, Massari F. An Insight on Novel Molecular Pathways in Metastatic Prostate Cancer: A Focus on DDR, MSI and AKT. Int J Mol Sci 2021; 22:ijms222413519. [PMID: 34948314 PMCID: PMC8708596 DOI: 10.3390/ijms222413519] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/07/2021] [Accepted: 12/15/2021] [Indexed: 02/06/2023] Open
Abstract
Prostate cancer is still one of the main causes of cancer-related death in the male population, regardless of the advancements in the treatment scenario. The genetic knowledge on prostate cancer is widely increasing, allowing researchers to identify novel promising molecular targets and treatment approaches. Genomic profiling has evidenced that DNA damage repair genes’ alterations are quite frequent in metastatic, castration resistant prostate cancer and specific therapies can interfere with this pathway, showing promising activity in this setting. Microsatellite instability is gaining attention as it seems to represent a predictive factor of the response to immunotherapy. Furthermore, the PTEN-PI3K-AKT pathway is another possible treatment target being investigated. In this review, we explore the current knowledge on these frequent genomic alterations of metastatic prostate cancer, their possible therapeutic repercussions and the promising future treatments under evaluation.
Collapse
Affiliation(s)
- Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni-15, 40138 Bologna, Italy; (V.M.); (A.M.); (M.R.); (G.N.); (A.R.); (F.M.)
| | - Andrea Marchetti
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni-15, 40138 Bologna, Italy; (V.M.); (A.M.); (M.R.); (G.N.); (A.R.); (F.M.)
| | - Matteo Rosellini
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni-15, 40138 Bologna, Italy; (V.M.); (A.M.); (M.R.); (G.N.); (A.R.); (F.M.)
| | - Giacomo Nuvola
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni-15, 40138 Bologna, Italy; (V.M.); (A.M.); (M.R.); (G.N.); (A.R.); (F.M.)
| | - Alessandro Rizzo
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni-15, 40138 Bologna, Italy; (V.M.); (A.M.); (M.R.); (G.N.); (A.R.); (F.M.)
| | - Matteo Santoni
- Oncology Unit, Macerata Hospital, 62100 Macerata, Italy;
| | - Alessia Cimadamore
- Section of Pathological Anatomy, School of Medicine, Polytechnic University of the Marche Region, United Hospitals, 60126 Ancona, Italy
- Correspondence:
| | - Rodolfo Montironi
- Molecular Medicine and Cell Therapy Foundation, Department of Clinical and Molecular Sciences, Polytechnic University of the Marche Region, 60100 Ancona, Italy;
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni-15, 40138 Bologna, Italy; (V.M.); (A.M.); (M.R.); (G.N.); (A.R.); (F.M.)
| |
Collapse
|
19
|
Jacob A, Raj R, Allison DB, Myint ZW. Androgen Receptor Signaling in Prostate Cancer and Therapeutic Strategies. Cancers (Basel) 2021; 13:5417. [PMID: 34771580 PMCID: PMC8582395 DOI: 10.3390/cancers13215417] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/21/2021] [Accepted: 10/27/2021] [Indexed: 11/17/2022] Open
Abstract
Understanding of the molecular mechanisms of prostate cancer has led to development of therapeutic strategies targeting androgen receptor (AR). These androgen-receptor signaling inhibitors (ARSI) include androgen synthesis inhibitor-abiraterone and androgen receptor antagonists-enzalutamide, apalutamide, and darolutamide. Although these medications provide significant improvement in survival among men with prostate cancer, drug resistance develops in nearly all patients with time. This could be through androgen-dependent or androgen-independent mechanisms. Even weaker signals and non-canonical steroid ligands can activate AR in the presence of truncated AR-splice variants, AR overexpression, or activating mutations in AR. AR splice variant, AR-V7 is the most studied among these and is not targeted by available ARSIs. Non-androgen receptor dependent resistance mechanisms are mediated by activation of an alternative signaling pathway when AR is inhibited. DNA repair pathway, PI3K/AKT/mTOR pathway, BRAF-MAPK and Wnt signaling pathway and activation by glucocorticoid receptors can restore downstream signaling in prostate cancer by alternative proteins. Multiple clinical trials are underway exploring therapeutic strategies to overcome these resistance mechanisms.
Collapse
Affiliation(s)
- Aasems Jacob
- Department of Medicine, Division of Hematology & Oncology, Pikeville Medical Center, Pikeville, KY 41501, USA;
| | - Rishi Raj
- Department of Medicine, Division of Endocrinology, Diabetes & Metabolism, Pikeville Medical Center, Pikeville, KY 41501, USA;
| | - Derek B. Allison
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA;
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY 40536, USA
- Department of Urology, University of Kentucky, Lexington, KY 40536, USA
| | - Zin W. Myint
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA;
- Department of Medicine, Division of Medical Oncology, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
20
|
Brady L, Carlsson J, Baird AM, Casey O, Vlajnic T, Murchan P, Cormican D, Costigan D, Gray S, Sheils O, O'Neill A, Watson RW, Andren O, Finn S. Correlation of integrated ERG/PTEN assessment with biochemical recurrence in prostate cancer. Cancer Treat Res Commun 2021; 29:100451. [PMID: 34507017 DOI: 10.1016/j.ctarc.2021.100451] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/19/2021] [Accepted: 08/29/2021] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Prostate cancer is a heterogeneous disease, with a complex molecular landscape that evolves throughout disease progression. Common alterations in genes such as ERG and PTEN have been attributed to worse prognosis. This study aimed to further examine the clinical relevance of PTEN and ERG expression in a cohort of patients with prostate cancer post radical prostatectomy. METHODS Tissue microarrays were constructed from 132 patients with prostate cancer from the Irish Prostate Cancer Research Consortium and University Hospital of Orebro, Sweden. Patients were divided into three groups - Group 1: biochemical recurrence, Group 2: no biochemical recurrence and Group 3: immediate progression after surgery. PTEN and ERG immunohistochemical analysis was performed and the association between expression levels and clinical parameters were compared. RESULTS Pathological stage pT3 tumours were more common at borderline significantly higher levels amongst patients who biochemically recurred when compared to patients who did not recur after radical prostatectomy (p = 0.05). ERG and PTEN expression levels were compared separately and concurrently across all three patient groups. Lack of ERG expression was strongly associated with immediate progression after surgery (p = 0.029). Loss of/low PTEN trended towards an association with immediate progression, however this was not statistically significant (p = 0.066). CONCLUSION In this study, negative ERG expression was strongly associated with immediate biochemical progression after radical prostatectomy. Moreover, a trend towards a relationship between aberrant PTEN expression and progression was observed. Additional studies with long-term follow up data may provide further clinical insight into the genomic heterogeneity in this population.
Collapse
Affiliation(s)
- Lauren Brady
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, Ireland
| | - Jessica Carlsson
- Department of Urology, Faculty of Medicine and Health, Örebro University, Orebro, Sweden
| | - Anne-Marie Baird
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Ireland
| | - Orla Casey
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, Ireland
| | - Tatjana Vlajnic
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Switzerland
| | - Pierre Murchan
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, Ireland
| | - David Cormican
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, Ireland
| | - Danielle Costigan
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, Ireland
| | - Steven Gray
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, Trinity College Dublin, Ireland
| | - Orla Sheils
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Ireland
| | - Amanda O'Neill
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - R William Watson
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Ove Andren
- Department of Urology, Faculty of Medicine and Health, Örebro University, Orebro, Sweden
| | - Stephen Finn
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, Ireland; Department of Histopathology, St James's Hospital, Dublin, Ireland.
| |
Collapse
|
21
|
Decreased ATM Protein Expression Is Substantiated with PTEN Loss in Defining Aggressive Phenotype of Prostate Cancer Associated with Lethal Disease. EUR UROL SUPPL 2021; 29:93-101. [PMID: 34337539 PMCID: PMC8317877 DOI: 10.1016/j.euros.2021.05.004] [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] [Accepted: 05/11/2021] [Indexed: 11/24/2022] Open
Abstract
Background Ataxia Telangiectasia Mutated (ATM) serine/threonine protein kinase is a known tumor suppressor, involved in DNA damage repair. It has prognostic and predictive therapeutic implications and is associated with aggressive prostate cancer (PCa). Objective To investigate the prognostic value of ATM protein expression in PCa patients and assessed the combined value of ATM, ERG, and PTEN status. Design, setting, and participants This study consisted of 303 patients with incidental, locally advanced, and castrate-resistant PCa by transurethral resection of the prostate (TURP). Outcome measurements and statistical analysis TURP samples from 303 PCa patients were assessed by immunohistochemistry (IHC for ATM, ERG, and PTEN. Individual and combined marker status were correlated with International Society of Urological Pathology Gleason grade group, overall survival (OS), and PCa-specific mortality (PCSM). Results and limitations Decreased ATM expression (negative/weak intensity) occurred in 164/303 (54.1%) patients, and was associated with shorter OS and higher PCSM (p = 0.015 and p = 0.001, respectively). Negative/weak ATM expression was significantly associated with PCSM with a hazard ratio of 2.09 (95% confidence interval 1.34–3.27, p = 0.001). Assessment of Combined ATM/PTEN expression showed improved prognostic power to predict OS and PCSM, independent of Gleason grade groups. Conclusions Decreased ATM protein expression is associated with poor outcomes in advanced PCa patients. Patients with combined low ATM/PTEN negative expression are at the highest risk for reduced OS and PCSM. Assessing the combined status of ATM/PTEN by IHC in PCa patients may aid in risk stratification relative to OS and PCSM. Moreover, since ATM plays an integral role in DNA damage response pathways, future studies will enhance our understanding of how outcomes of patients with altered ATM and PTEN expression can be improved further with poly-ADP ribose polymerase inhibitors (PARPi), combinations of PARPi and androgen receptor–targeted therapies, as well as platinum-based chemotherapies. Patient summary Lower ATM intensity is associated with increased cancer-specific mortality in prostate cancer patients. Patients with lower ATM and PTEN negative expression showed decreased overall survival and increased cancer mortality compared with controls.
Collapse
|
22
|
Danesh Pazhooh R, Rahnamay Farnood P, Asemi Z, Mirsafaei L, Yousefi B, Mirzaei H. mTOR pathway and DNA damage response: A therapeutic strategy in cancer therapy. DNA Repair (Amst) 2021; 104:103142. [PMID: 34102579 DOI: 10.1016/j.dnarep.2021.103142] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/26/2021] [Accepted: 05/31/2021] [Indexed: 10/21/2022]
Abstract
The mammalian target of rapamycin (mTOR) is a conserved serine/threonine-protein kinase, comprising two subunit protein complexes: mTORC1 and mTORC2. In response to insult and cancer, the mTOR pathway plays a crucial role in regulating growth, metabolism, cell survival, and protein synthesis. Key subunits of mTORC1/2 catalyze the phosphorylation of various molecules, including eukaryotic translation initiation factor 4E binding protein 1 (4E-BP1), ribosomal protein S6 kinase β-1 (S6K1). The DNA damage response (DDR) maintains genomic stability and provides an opportunity for treating tumors with defects caused by DNA damaging agents. Many mTOR inhibitors are utilized for the treatment of cancers. However, several clinical trials are still assessing the efficacy of mTOR inhibitors. This paper discusses the role of the mTOR signaling pathway and its regulators in developing cancer. In the following, we will review the interaction between DDR and mTOR signaling and the innovative therapies applied in preclinical and clinical trials for treating cancers.
Collapse
Affiliation(s)
| | | | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| | - Liaosadat Mirsafaei
- Department of Cardiology, Ramsar Campus, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
23
|
Salles DC, Vidotto T, Faisal FA, Tosoian JJ, Guedes LB, Muranyi A, Bai I, Singh S, Yan D, Shanmugam K, Lotan TL. Assessment of MYC/PTEN Status by Gene-Protein Assay in Grade Group 2 Prostate Biopsies. J Mol Diagn 2021; 23:1030-1041. [PMID: 34062284 PMCID: PMC8491088 DOI: 10.1016/j.jmoldx.2021.05.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/20/2021] [Accepted: 05/14/2021] [Indexed: 11/19/2022] Open
Abstract
This study leveraged a gene-protein assay to assess MYC and PTEN status at prostate cancer biopsy and examined the association with adverse outcomes after surgery. MYC gain and PTEN loss were simultaneously assessed by chromogenic in situ hybridization and immunohistochemistry, respectively, using 277 Grade Group 2 needle biopsies that were followed by prostatectomy. The maximal size of cribriform Gleason pattern 4 carcinoma (CRIB), the presence of intraductal carcinoma (IDC), and percentage of Gleason pattern 4 carcinoma at biopsy were also annotated. MYC gain or PTEN loss was present in 19% and 18% of biopsies, respectively, whereas both alterations were present in 9% of biopsies. Tumors with one or both alterations were significantly more likely to have non-organ-confined disease (NOCD) at radical prostatectomy. In logistic regression models, including clinical stage, tumor volume on biopsy, and presence of CRIB/IDC, cases with MYC gain and PTEN loss remained at higher risk for NOCD (odds ratio, 6.23; 95% CI, 1.74-24.55; P = 0.005). The area under the curve for a baseline model using CAPRA variables (age, prostate-specific antigen, percentage of core involvement, clinical stage) was increased from 0.68 to 0.69 with inclusion of CRIB/IDC status and to 0.75 with MYC/PTEN status. Dual MYC/PTEN status can be assessed in a single slide and is independently associated with increased risk of NOCD for Grade Group 2 biopsies.
Collapse
Affiliation(s)
- Daniela C Salles
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Thiago Vidotto
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Farzana A Faisal
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Liana B Guedes
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Isaac Bai
- Roche Tissue Diagnostics, Tucson, Arizona
| | | | | | | | - Tamara L Lotan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
24
|
Swellam M, Saad EA, Sabry S, Denewer A, Abdel Malak C, Abouzid A. Alterations of PTEN and SMAD4 methylation in diagnosis of breast cancer: implications of methyl II PCR assay. J Genet Eng Biotechnol 2021; 19:54. [PMID: 33825073 PMCID: PMC8024427 DOI: 10.1186/s43141-021-00154-x] [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: 12/21/2020] [Accepted: 03/26/2021] [Indexed: 12/22/2022]
Abstract
Background Diagnosis of breast cancer is more complicated due to lack of minimal invasive biomarker with sufficient precision. DNA methylation is a promising marker for cancer diagnosis. In this study, authors evaluated methylation patterns for PTEN and SMAD4 in blood samples using EpiTect Methyl II QPCR assay quantitative PCR technology. Results Methylation status for PTEN and SMAD4 were statistically significant as breast cancer patients reported hypermethylation compared to benign and control groups (77.1 ± 17.9 vs. 24.9 ± 4.5 and 15.1 ± 1.4 and 70.1 ± 14.4 vs. 28.2 ± 0.61 and 29.5 ± 3.6, respectively). ROC curve analysis revealed that both PTEN (AUC = 0.992) and SMAD4 (AUC = 0.853) had good discriminative power for differentiating BC from all non-cancer individuals (benign and healthy combined) compared to routine tumor markers CEA (AUC = 0.538) and CA15.3 (AUC = 0.686). High PTEN methylation degree was associated with late stages (84.2 ± 17.4), positive lymph node (84.2 ± 18.5), positive ER (81.3 ± 19.7), positive PgR (79.5 ± 19.1), and positive HER2 (80.7 ± 19.0) vs. 67.4 ± 13.8, 70.6 ± 14.8, 72.8 ± 14.9, 72.5 ± 14.7, and 70.2 ± 13.5 in early stages, negative lymph node, negative ER, negative PgR, and negative HER2, respectively. Similar results were obtained regarding SMAD4 methylation. Sensitivity, specificity, positive and negative predictive values, and accuracy for methylated PTEN were 100%, 95%, 99.1%, 100%, and 95%, respectively when differentiated BC from all-non cancer controls. Interestingly, PTEN could distinguish early BC stages with good sensitivity 84.4%, 51.4%, 69.1%, 72%, and 70%, respectively. Conclusion Methylation status of PTEN and SMAD4 is a promising blood marker for early detection of breast cancer. Future studies are needed for their role as prognostic markers.
Collapse
Affiliation(s)
- Menha Swellam
- Biochemistry Department, Genetic Engineering and Biotechnology Research Division, High Throughput Molecular and Genetic Laboratory, Center for Excellences for Advanced Sciences, National Research Centre, Dokki, Giza, Egypt
| | - Entsar A Saad
- Chemistry Department, Faculty of Science, Damietta University, Damietta, 34517, Egypt
| | - Shimaa Sabry
- Chemistry Department, Faculty of Science, Damietta University, Damietta, 34517, Egypt.
| | - Adel Denewer
- Surgical Oncology Department, Mansoura Oncology Centre, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Camelia Abdel Malak
- Chemistry Department, Faculty of Science, Damietta University, Damietta, 34517, Egypt
| | - Amr Abouzid
- Surgical Oncology Department, Mansoura Oncology Centre, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
25
|
Conteduca V, Mosca A, Brighi N, de Giorgi U, Rescigno P. New Prognostic Biomarkers in Metastatic Castration-Resistant Prostate Cancer. Cells 2021; 10:193. [PMID: 33478015 PMCID: PMC7835961 DOI: 10.3390/cells10010193] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 12/13/2022] Open
Abstract
Prostate cancer is one of the most frequent cancers in men and is a common cause of cancer-related death. Despite significant progress in the diagnosis and treatment of this tumor, patients who relapse after radical treatments inevitably develop metastatic disease. Patient stratification is therefore key in this type of cancer, and there is an urgent need for prognostic biomarkers that can define patients' risk of cancer-related death. In the last 10 years, multiple prognostic factors have been identified and studied. Here, we review the literature available and discuss the most common aberrant genomic pathways in metastatic castration-resistant prostate cancer shown to have a prognostic relevance in this setting.
Collapse
Affiliation(s)
- Vincenza Conteduca
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (V.C.); (N.B.); (U.d.G.)
| | - Alessandra Mosca
- Multidisciplinary Outpatient Oncology Clinic, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060 Turin, Italy;
| | - Nicole Brighi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (V.C.); (N.B.); (U.d.G.)
| | - Ugo de Giorgi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (V.C.); (N.B.); (U.d.G.)
| | - Pasquale Rescigno
- Interdisciplinary Group for Translational Research and Clinical Trials, Urological Cancers (GIRT-Uro), Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060 Turin, Italy
| |
Collapse
|
26
|
Abou-Ouf H, Assem H, Ghosh S, Karnes RJ, Stoletov K, Palanisamy N, Lewis JD, Bismar TA. High Serine-arginine Protein Kinase 1 Expression with PTEN Loss Defines Aggressive Phenotype of Prostate Cancer Associated with Lethal Outcome and Decreased Overall Survival. EUR UROL SUPPL 2020; 23:1-8. [PMID: 34337483 PMCID: PMC8317848 DOI: 10.1016/j.euros.2020.11.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
Background Serine-arginine protein kinase 1 (SRPK1) has been implicated in prostate cancer (PCa) progression. However, its prognostic value and association with ERG and PTEN expression, two of the most common genetic alterations, have not been explored fully. Objective We assessed the prognostic value of SRPK1 in association with ERG and PTEN in a cohort of patients managed nonsurgically by androgen deprivation therapy (ADT) for advanced disease. Design, setting, and participants The study cohort consisted of men diagnosed with PCa by transurethral resection of the prostate (TURP; n = 480). The patients were divided into three main groups: incidental (patients with Gleason score [GS] ≤7 with no prior ADT), advanced (patients with GS ≥8 with no prior ADT), and castrate-resistant PCa (patients with prior ADT). Outcome measurements and statistical analysis A total of 480 TURP samples were assessed by immunohistochemistry for SRPK1, ERG, and PTEN, and results were correlated with Gleason grade group (GG), overall survival (OS), and PCa-specific mortality (PCSM). Results and limitations High SRPK1 expression was noted in 105/455 (23%) available patient cores. Expression of SRPK1 was associated with Gleason grade grouping (p < 0.0001) with high expression detected in 22/74 (33%) with GG 5. High SRPK1 was not associated with ERG positivity (p = 0.18) but was significantly associated with PTEN intensity (p = 0.001). High SRPK1 was associated with OS (hazard ratio [HR] 1.99; confidence interval [CI]: 1.57–2.54, p < 0.0001) and PCSM (HR 1.64; CI: 1.19–2.26, p < 0.002). Adjusting for Gleason score, patients with high SRPK1 and negative PTEN had the worst clinical outcome for both OS and PCSM compared with other patients (p < 0.0001, HR: 3.02; CI: 1.87–4.88 and HR: 6.40, CI: 3.19–12.85, respectively). Conclusions High SRPK1 is associated with worse OS and PCSM. Moreover, patients with high SRPK1 expression and loss of PTEN had the worst clinical outcome for OS and cancer-specific mortality. Combined status of SRPK1 and PTEN may provide added value in stratifying patients into various prognostic groups. Patient summary The expression of serine-arginine protein kinase 1 (SRPK1) combined with PTEN has a significant prognostic role in prostate cancer patients. Patients with high SRPK1 expression and negative PTEN had the worst clinical outcome for overall survival and cancer-specific mortality.
Collapse
Affiliation(s)
- Hatem Abou-Ouf
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine and Alberta Public Laboratories, University of Calgary, Calgary, Alberta, Canada
| | - Hisham Assem
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine and Alberta Public Laboratories, University of Calgary, Calgary, Alberta, Canada
| | - Sunita Ghosh
- Department of Medical Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.,Alberta Health Services-Cancer Control, Edmonton, Alberta, Canada
| | | | - Konstantin Stoletov
- Department of Medical Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Nallasivam Palanisamy
- Department of Urology, Vattikuti Urology Institute, Henry Ford Health System Detroit, MI, USA
| | - John D Lewis
- Department of Medical Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Tarek A Bismar
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine and Alberta Public Laboratories, University of Calgary, Calgary, Alberta, Canada.,Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Arnie Charbonneau Cancer Institute, Tom Baker Cancer Center and Alberta Public Laboratories, Calgary, Alberta, Canada
| |
Collapse
|
27
|
Vlajnic T, Bubendorf L. Molecular pathology of prostate cancer: a practical approach. Pathology 2020; 53:36-43. [PMID: 33234230 DOI: 10.1016/j.pathol.2020.10.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 10/14/2020] [Indexed: 12/13/2022]
Abstract
While localised prostate cancer can be cured by local treatment, 'high-risk' prostate cancer often progresses to castration resistant disease and remains incurable with a dismal prognosis. In recent years, technical advances and development of novel methodologies have largely contributed to a better understanding of underlying molecular mechanisms that promote tumour growth and progression. Consecutively, novel therapeutic strategies for treatment of prostate cancer have emerged during the last decade, calling for the identification of predictive biomarkers. The concept of personalised medicine is to tailor treatment according to the specific tumour profile of an individual patient. Moreover, acquired molecular changes during tumour evolution and in response to therapy selection pressure require adapted predictive marker testing at different time points during the disease. In this setting, the pathologist plays a critical role in patient management and treatment selection. In this review, we provide a comprehensive overview of the current knowledge of molecular aspects of prostate cancer and their potential utility in the context of different therapeutic approaches. Furthermore, we discuss methods for molecular marker testing in routine clinical practice, with a focus on castration resistant prostate cancer.
Collapse
Affiliation(s)
- Tatjana Vlajnic
- Institute of Pathology, University Hospital Basel, Basel, Switzerland.
| | - Lukas Bubendorf
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
28
|
da Silva FC, Algaba F. Re: Report from the International Society of Urological Pathology (ISUP) Consultation Conference on Molecular Pathology of Urogenital Cancers. I. Molecular Biomarkers in Prostate Cancer. Eur Urol 2020; 79:318-319. [PMID: 33218825 DOI: 10.1016/j.eururo.2020.10.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 10/22/2020] [Indexed: 10/23/2022]
Affiliation(s)
| | - Ferran Algaba
- Section of Pathology, Fundació Puigvert, Universitat Autónoma de Barcelona, Barcelona, Spain
| |
Collapse
|
29
|
Report From the International Society of Urological Pathology (ISUP) Consultation Conference on Molecular Pathology of Urogenital Cancers. I. Molecular Biomarkers in Prostate Cancer. Am J Surg Pathol 2020; 44:e15-e29. [PMID: 32044806 DOI: 10.1097/pas.0000000000001450] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The combined clinical and molecular heterogeneity of prostate cancer necessitates the use of prognostic, predictive, and diagnostic biomarkers to assist the clinician with treatment selection. The pathologist plays a critical role in guiding molecular biomarker testing in prostate cancer and requires a thorough knowledge of the current testing options. In the setting of clinically localized prostate cancer, prognostic biomarkers such as Ki-67 labeling, PTEN loss or mRNA-based genomic signatures can be useful to help determine whether definitive therapy is required. In the setting of advanced disease, predictive biomarkers, such as the presence of DNA repair deficiency mediated by BRCA2 loss or mismatch repair gene defects, may suggest the utility of poly-ADP ribosylase inhibition or immune checkpoint blockade. Finally, androgen receptor-related biomarkers or diagnostic biomarkers indicating the presence of small cell neuroendocrine prostate cancer may help guide the use of androgen receptor signaling inhibitors and chemotherapy. In this review, we examine the current evidence for several prognostic, predictive and diagnostic tissue-based molecular biomarkers in prostate cancer management. For each assay, we summarize a recent survey of the International Society of Urology Pathology (ISUP) members on current testing practices and include recommendations for testing that emerged from the ISUP Working Group on Molecular Pathology of Prostate Cancer and the 2019 Consultation Conference on Molecular Pathology of Urogenital Cancers.
Collapse
|
30
|
Liu W, Hou J, Petkewicz J, Na R, Wang CH, Sun J, Gallagher J, Bogachkov YY, Swenson L, Regner M, Resurreccion WK, Isaacs WB, Brendler CB, Crawford S, Zheng SL, Helfand BT, Xu J. Feasibility and performance of a novel probe panel to detect somatic DNA copy number alterations in clinical specimens for predicting prostate cancer progression. Prostate 2020; 80:1253-1262. [PMID: 32803894 DOI: 10.1002/pros.24057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 08/03/2020] [Indexed: 11/11/2022]
Abstract
BACKGROUND To assess the feasibility of a novel DNA-based probe panel to detect copy number alterations (CNAs) in prostate tumor DNA and its performance for predicting clinical progression. METHODS A probe panel was developed and optimized to measure CNAs in trace amounts of tumor DNA (2 ng) isolated from formalin-fixed paraffin-embedded tissues. Ten genes previously associated with aggressive disease were targeted. The panel's feasibility and performance were assessed in 175 prostate cancer (PCa) patients who underwent radical prostatectomy with a median 10-year follow-up, including 42 men who developed disease progression (either metastasis and/or PCa-specific death). Association with disease progression was tested using univariable and multivariable analyses. RESULTS The probe panel detected CNAs in all 10 genes in tumor DNA isolated from either diagnostic biopsies or surgical specimens. A four-gene model (PTEN/MYC/BRCA2/CDKN1B) had the strongest association with disease progression; 64.3% of progressors and 22.5% of non-progressors had at least one CNA in these four genes, odds ratio (OR) (95% confidence interval) = 6.21 (2.77-13.87), P = 8.48E-06. The association with disease progression remained significant after adjusting for known clinicopathological variables. Among the seven progressors of the 65 patients with clinically low-risk disease, three (42.9%) had at least one CNA in these four genes. CONCLUSIONS The probe panel can detect CNAs in trace amounts of tumor DNA from biopsies or surgical tissues at the time of diagnosis or surgery. CNAs independently predict metastatic/lethal cancer, particularly among men with clinically low-risk disease at diagnosis. If validated, this may improve current abilities to assess tumor aggressiveness.
Collapse
Affiliation(s)
- Wennuan Liu
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois
| | - Jun Hou
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
| | - Jacqueline Petkewicz
- John and Carol Walter Center for Urological Health, NorthShore University HealthSystem, Evanston, Illinois
| | - Rong Na
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
| | - Chi-Hsiung Wang
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois
| | - Jishan Sun
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois
| | - Johnie Gallagher
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
| | - Yedida Y Bogachkov
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
| | - Laura Swenson
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
| | - MaryAnn Regner
- Department of Pathology, NorthShore University HealthSystem, Evanston, Illinois
| | - W Kyle Resurreccion
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
| | - William B Isaacs
- Department of Urology and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Charles B Brendler
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois
| | - Susan Crawford
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois
| | - S Lilly Zheng
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois
| | - Brian T Helfand
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois
- John and Carol Walter Center for Urological Health, NorthShore University HealthSystem, Evanston, Illinois
| | - Jianfeng Xu
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois
| |
Collapse
|
31
|
Hasan S. An Overview of Promising Biomarkers in Cancer Screening and Detection. Curr Cancer Drug Targets 2020; 20:831-852. [PMID: 32838718 DOI: 10.2174/1568009620666200824102418] [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: 05/13/2020] [Revised: 07/03/2020] [Accepted: 07/13/2020] [Indexed: 11/22/2022]
Abstract
Applications of biomarkers have been proved in oncology screening, diagnosis, predicting response to treatment as well as monitoring the progress of the disease. Considering the crucial role played by them during different disease stages, it is extremely important to evaluate, validate, and assess them to incorporate them into routine clinical care. In this review, the role of few most promising and successfully used biomarkers in cancer detection, i.e. PD-L1, E-Cadherin, TP53, Exosomes, cfDNA, EGFR, mTOR with regard to their structure, mode of action, and reports signifying their pathological significance, are addressed. Also, an overview of some successfully used biomarkers for cancer medicine has been presented. The study also summarizes biomarker-driven personalized cancer therapy i.e., approved targets and indications, as per the US FDA. The review also highlights the increasingly prominent role of biomarkers in drug development at all stages, with particular reference to clinical trials. The increasing utility of biomarkers in clinical trials is clearly evident from the trend shown, wherein ~55 percent of all oncology clinical trials in 2019 were seen to involve biomarkers, as opposed to ~ 15 percent in 2001, which clearly proves the essence and applicability of biomarkers for synergizing clinical information with tumor progression. Still, there are significant challenges in the implementation of these possibilities with strong evidence in cost-- effective manner.
Collapse
Affiliation(s)
- Saba Hasan
- Amity Institute of Biotechnology, Amity University, Uttar Pradesh, Lucknow, India
| |
Collapse
|
32
|
Shin SH, Kim I, Lee JE, Lee M, Park JW. Loss of EGR3 is an independent risk factor for metastatic progression in prostate cancer. Oncogene 2020; 39:5839-5854. [PMID: 32796959 DOI: 10.1038/s41388-020-01418-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 07/29/2020] [Accepted: 08/04/2020] [Indexed: 12/14/2022]
Abstract
Identification of pro-metastatic genomic alterations is urgently needed to help understand and prevent the fatal course of prostate cancer. Here, we found that the transcription factor EGR3, located at chromosome 8p21.3, is a critical metastasis suppressor. Aberrant deletion of EGR3 was found in up to 59.76% (deep deletions, 16.87%; shallow deletions, 42.89%) of prostate cancer patients. In informatics analysis, EGR3 loss was associated with prostate cancer progression and low survival rates. EGR3 expression inversely correlated with the expressions of epithelial-to-mesenchymal transition (EMT) and metastasis-related gene sets in prostate cancer tissues. In prostate cancer cells, EGR3 blocked the EMT process and suppressed cell migration and invasion. In a mouse model for cancer metastasis, EGR3 overexpression significantly suppressed bone metastases of PC3 and 22Rv1 prostate cancer cells. Mechanistically, EGR3 transcriptionally activated ZFP36, GADD45B, and SOCS3 genes by directly binding to their promoter regions. The EMT-inhibitory and tumor-suppressive roles of the EGR3 downstream genes were identified through in vitro and in silico analyses. Together, our results showed that EGR3 may be a biomarker to predict clinical outcomes and that it plays an important role in the metastatic progression of prostate cancer.
Collapse
Affiliation(s)
- Seung-Hyun Shin
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,Department of Pharmacology, Seoul National University College of Medicine, Seoul, South Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Iljin Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,Department of Pharmacology, Seoul National University College of Medicine, Seoul, South Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea.,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Jae Eun Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,Department of Pharmacology, Seoul National University College of Medicine, Seoul, South Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Mingyu Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,Department of Pharmacology, Seoul National University College of Medicine, Seoul, South Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea.,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, South Korea.,Obstructive Upper airway Research (OUaR) Laboratory, Seoul National University College of Medicine, Seoul, South Korea
| | - Jong-Wan Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea. .,Department of Pharmacology, Seoul National University College of Medicine, Seoul, South Korea. .,Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea. .,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, South Korea.
| |
Collapse
|
33
|
Kaczorowski A, Tolstov Y, Falkenstein M, Vasioukhin V, Prigge ES, Geisler C, Kippenberger M, Nientiedt C, Ratz L, Kuryshev V, Herpel E, Kristiansen G, Sültmann H, Stenzinger A, Doeberitz MVK, Hohenfellner M, Duensing A, Duensing S. Rearranged ERG confers robustness to prostate cancer cells by subverting the function of p53. Urol Oncol 2020; 38:736.e1-736.e10. [PMID: 32674955 DOI: 10.1016/j.urolonc.2020.06.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/06/2020] [Accepted: 06/15/2020] [Indexed: 12/25/2022]
Abstract
OBJECTIVE ERG rearrangements are frequent and early events in prostate cancer. The functional role of rearranged ERG, however, is still incompletely understood. ERG rearrangements are maintained during prostate cancer progression suggesting that they may confer a selective advantage. The molecular basis of this notion is the subject of this study. METHODS A variety of immunological methods were used to characterize the effects of rearranged ERG on p53. Consequences of an overexpression of N-terminally deleted ERG on p53 function were interrogated by measuring apoptosis and cellular senescence in the presence or absence of exogenous DNA damage. Effects of N-terminally deleted ERG on the transactivation function of p53 were analyzed by qRT-PCR. RESULTS We show that overexpression of ERG leads to an increased basal level of DNA damage and a stabilization of p53 that involves a sequestration of its E3 ubiquitin ligase, MDM2, into nucleoli. A higher p53 expression was also observed in vivo in an ERG-overexpressing prostatic intraepithelial neoplasia mouse model. The correlation between ERG and p53 expression was corroborated in 163 patients with prostate cancer. ERG overexpression was found to inhibit both apoptosis and cellular senescence induced by exogenous DNA damage. Mechanistically, this protective effect of ERG involved an abrogation of the DNA damage-induced expression of p53 target genes. CONCLUSIONS By protecting tumor cells from the antiproliferative consequences of genotoxic stress, ERG may allow the survival and proliferation of genomically unstable tumor cells. Targeting ERG may therefore represent a promising strategy to suppress such adverse features during prostate cancer progression.
Collapse
Affiliation(s)
- Adam Kaczorowski
- Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 517, D-69120 Heidelberg, Germany
| | - Yanis Tolstov
- Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 517, D-69120 Heidelberg, Germany
| | - Michael Falkenstein
- Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 517, D-69120 Heidelberg, Germany
| | - Valeri Vasioukhin
- Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview, Avenue N C3-168, Seattle, 98109, Washington
| | - Elena-Sophie Prigge
- Department of Applied Tumor Biology, Institute of Pathology, University Hospital, Heidelberg, and Clinical Cooperation Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 224, D-69120 Heidelberg, Germany
| | - Christine Geisler
- Department of Urology, University Hospital Heidelberg, and National Center for Tumor Diseases (NCT) Heidelberg, Im Neuenheimer Feld 110, D-69120 Heidelberg, Germany
| | - Maximilian Kippenberger
- Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 517, D-69120 Heidelberg, Germany
| | - Cathleen Nientiedt
- Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 517, D-69120 Heidelberg, Germany; Department of Medical Oncology, University Hospital Heidelberg, National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, D-69120 Heidelberg, Germany
| | - Leonie Ratz
- Cancer Genome Research, National Center for Tumor Diseases (NCT) Heidelberg, German Cancer Research Center (DKFZ), and German Cancer Consortium (DKTK), Im Neuenheimer Feld 460, D-69120 Heidelberg, Germany
| | - Vladimir Kuryshev
- Cancer Genome Research, National Center for Tumor Diseases (NCT) Heidelberg, German Cancer Research Center (DKFZ), and German Cancer Consortium (DKTK), Im Neuenheimer Feld 460, D-69120 Heidelberg, Germany
| | - Esther Herpel
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, D-69120, Heidelberg, Germany; Tissue Bank of the National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 224, D-69120 Heidelberg, Germany
| | - Glen Kristiansen
- Institute of Pathology, University Hospital Bonn, Sigmund-Freud-Strasse 25, D-53127 Bonn, Germany
| | - Holger Sültmann
- Cancer Genome Research, National Center for Tumor Diseases (NCT) Heidelberg, German Cancer Research Center (DKFZ), and German Cancer Consortium (DKTK), Im Neuenheimer Feld 460, D-69120 Heidelberg, Germany
| | - Albrecht Stenzinger
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, D-69120, Heidelberg, Germany
| | - Magnus von Knebel Doeberitz
- Department of Applied Tumor Biology, Institute of Pathology, University Hospital, Heidelberg, and Clinical Cooperation Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 224, D-69120 Heidelberg, Germany
| | - Markus Hohenfellner
- Department of Urology, University Hospital Heidelberg, and National Center for Tumor Diseases (NCT) Heidelberg, Im Neuenheimer Feld 110, D-69120 Heidelberg, Germany
| | - Anette Duensing
- Department of Urology, University Hospital Heidelberg, and National Center for Tumor Diseases (NCT) Heidelberg, Im Neuenheimer Feld 110, D-69120 Heidelberg, Germany; Precision Oncology of Urological Malignancies, Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 517, D-69120 Heidelberg, Germany; Cancer Therapeutics Program, UPMC Hillman Cancer Center, 5117 Centre Avenue, Pittsburgh, 15213, Pennsylvania; Department of Pathology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, 15213, Pennsylvania
| | - Stefan Duensing
- Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 517, D-69120 Heidelberg, Germany; Department of Urology, University Hospital Heidelberg, and National Center for Tumor Diseases (NCT) Heidelberg, Im Neuenheimer Feld 110, D-69120 Heidelberg, Germany.
| |
Collapse
|
34
|
The triphenyltin carboxylate derivative triphenylstannyl 2-(benzylcarbamoyl)benzoate impedes prostate cancer progression via modulation of Akt/FOXO3a signaling. Toxicol Appl Pharmacol 2020; 401:115091. [PMID: 32525019 DOI: 10.1016/j.taap.2020.115091] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 05/31/2020] [Accepted: 06/02/2020] [Indexed: 12/22/2022]
Abstract
Prostate cancer (PCa) incidence is surging in United States and other parts of the world. Synthetic and natural compounds have been explored as potential modulators of PI3K/Akt signaling that is known to drive PCa growth. Here, we evaluated the efficacy of a series of triphenyltin (IV) carboxylate derivatives against PCa. From this library, triphenylstannyl 2-(benzylcarbamoyl)benzoate (Ch-319) resulted in reduced viability and induction of cell cycle arrest in PTEN-/- PC3M and PTEN+/- DU145 cells. In parallel, downregulation of PI3K p85/p110 subunits, dephosphorylation of Akt-1 and increase in FOXO3a expression were observed. In silico studies indicated binding interactions of Ch-319 within the ATP binding site of Akt-1 at Met281, Phe442 and Glu234 residues. Elevated po-pulation of apoptotic cells, activation of Bax and reduced Bcl2 expression indicated apoptosis by Ch-319. Pre-sensitization of PCa cells with Ch-319 augmented the effect of cabazitaxel, a commonly used taxane in patients with castration-resistant PCa. Next, in a prostate-specific PTENp-/- mice, Ch-319 showed reduced weights of genitourinary apparatus as compared to DMSO treated controls. Histological studies indicated absence of neoplastic foci in Ch-319 treated prostates. Consistently, dephosphorylation of Akt-1, reduced expression of PRAS40 and androgen receptor and increase in FOXO3a were observed in treated group. Notably, no overt organ toxicity was noted in Ch-319 treated animals. Our studies identify Akt/FOXO3a signaling as a target of triphenyltin (IV) carboxylate Ch-319 and provide a molecular basis of its growth inhibitory effect in PCa cells. We propose that Ch-319 has the potential to be developed as an anticancer agent against PCa.
Collapse
|
35
|
Hashim D, Gonzalez-Feliciano AG, Ahearn TU, Pettersson A, Barber L, Pernar CH, Ebot EM, Isikbay M, Finn SP, Giovannucci EL, Lis RT, Loda M, Parmigiani G, Lotan T, Kantoff PW, Mucci LA, Graff RE. Family history of prostate cancer and the incidence of ERG- and phosphatase and tensin homolog-defined prostate cancer. Int J Cancer 2020; 146:2694-2702. [PMID: 31318977 DOI: 10.1002/ijc.32577] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 06/18/2019] [Accepted: 06/28/2019] [Indexed: 01/08/2023]
Abstract
Family history is among the strongest known risk factors for prostate cancer (PCa). Emerging data suggest molecular subtypes of PCa, including two somatic genetic aberrations: fusions of androgen-regulated promoters with ERG and, separately, phosphatase and tensin homolog (PTEN) loss. We examined associations between family history and incidence of these subtypes in 44,126 men from the prospective Health Professionals Follow-up Study. ERG and PTEN status were assessed by immunohistochemistry. Multivariable competing risks models were used to estimate hazard ratios (HR) and 95% confidence intervals (CI) for associations between self-reported family history of PCa and molecular subtypes of disease. Thirteen percent of men had a positive family history of PCa at baseline. During a median follow-up of 18.5 years, 5,511 PCa cases were diagnosed. Among them, 888 were assayed for ERG status (47% ERG-positive) and 715 were assayed for PTEN loss (14% PTEN null). Family history was more strongly associated with risk of ERG-negative (HR: 2.15; 95% CI: 1.71-2.70) than ERG-positive (HR: 1.49; 95% CI: 1.13-1.95) disease (pheterogeneity : 0.04). The strongest difference was among men with an affected father (HRERG-negative : 2.09; 95% CI: 1.64-2.66; HRERG-positive : 1.30; 95% CI: 0.96-1.76; pheterogeneity : 0.01). Family history of PCa was positively associated with both PTEN null (HR: 2.10; 95% CI: 1.26-3.49) and PTEN intact (HR: 1.72; 95% CI: 1.39-2.13) PCa (pheterogeneity : 0.47). Our results indicate that PCa family history may be positively associated with PCa in all ERG and PTEN subtypes, suggesting a role of genetic susceptibility in their development. It is possible that ERG-negative disease could be especially associated with positive family history.
Collapse
Affiliation(s)
- Dana Hashim
- Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY
| | | | - Thomas U Ahearn
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA.,Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Andreas Pettersson
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA.,Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Lauren Barber
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Claire H Pernar
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Ericka M Ebot
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Masis Isikbay
- Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - Stephen P Finn
- Department of Histopathology, St. James's Hospital and Trinity College Dublin Medical School, Dublin, Ireland
| | - Edward L Giovannucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA.,Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Rosina T Lis
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Massimo Loda
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Giovanni Parmigiani
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA
| | - Tamara Lotan
- Department of Pathology, Johns Hopkins Bayview Medical Center, Baltimore, MD
| | - Philip W Kantoff
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA.,Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Rebecca E Graff
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
36
|
Carceles-Cordon M, Kelly WK, Gomella L, Knudsen KE, Rodriguez-Bravo V, Domingo-Domenech J. Cellular rewiring in lethal prostate cancer: the architect of drug resistance. Nat Rev Urol 2020; 17:292-307. [PMID: 32203305 PMCID: PMC7218925 DOI: 10.1038/s41585-020-0298-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2020] [Indexed: 12/14/2022]
Abstract
Over the past 5 years, the advent of combination therapeutic strategies has substantially reshaped the clinical management of patients with advanced prostate cancer. However, most of these combination regimens were developed empirically and, despite offering survival benefits, are not enough to halt disease progression. Thus, the development of effective therapeutic strategies that target the mechanisms involved in the acquisition of drug resistance and improve clinical trial design are an unmet clinical need. In this context, we hypothesize that the tumour engineers a dynamic response through the process of cellular rewiring, in which it adapts to the therapy used and develops mechanisms of drug resistance via downstream signalling of key regulatory cascades such as the androgen receptor, PI3K-AKT or GATA2-dependent pathways, as well as initiation of biological processes to revert tumour cells to undifferentiated aggressive states via phenotype switching towards a neuroendocrine phenotype or acquisition of stem-like properties. These dynamic responses are specific for each patient and could be responsible for treatment failure despite multi-target approaches. Understanding the common stages of these cellular rewiring mechanisms to gain a new perspective on the molecular underpinnings of drug resistance might help formulate novel combination therapeutic regimens.
Collapse
Affiliation(s)
- Marc Carceles-Cordon
- Medical Oncology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - W Kevin Kelly
- Medical Oncology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Leonard Gomella
- Urology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Karen E Knudsen
- Medical Oncology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
- Urology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
- Cancer Biology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Veronica Rodriguez-Bravo
- Cancer Biology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| | - Josep Domingo-Domenech
- Medical Oncology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
- Cancer Biology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
37
|
Abdelsalam RA, Khalifeh I, Box A, Kalantarian M, Ghosh S, Abou-Ouf H, Lotfi T, Shahait M, Palanisamy N, Bismar TA. Molecular characterization of prostate cancer in Middle Eastern population highlights differences with Western populations with prognostic implication. J Cancer Res Clin Oncol 2020; 146:1701-1709. [PMID: 32350606 DOI: 10.1007/s00432-020-03221-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 04/17/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND To investigate the incidence and prognostication of ERG, PTEN and SPINK1 protein expressions in prostate cancer cohort of Middle Eastern descent in comparison to published data from Western population. METHODS Immunohistochemistry for ERG, PTEN and SPINK1 was performed in a cohort of localized PCA (n = 340). The data were correlated to pathological and clinical outcomes and compared to Western populations. RESULTS ERG expression and PTEN loss were noted in 123/288 (42.7%) and 91/297 (30.6%) of patients, respectively. SPINK1 expression was assessed in a subset of cases, noted in 6/150 (4%) of patients. Only ERG expression was associated with grade groups, being more common in the lower grade groups (1-3 vs 4-5; p = 0.04). In contrast to the Western population, PTEN loss foci were more likely to be ERG negative, observed in 81% of tumor foci and patients with PTEN neg/ERG pos were more likely to exhibit biochemical recurrence (OR 2.831; 95% CI 1.10-726, p = 0.03). This association remained significant in multivariate analysis (OR 2.68; 95% CI 0.98-7.33, p = 0.05), after adjusting for GG, path stage and surgical margin. CONCLUSION This study documents significant differences in key molecular events in PCA in Middle Eastern population compared to Western populations that could explain differences in PCA incidence, progression and prognostication. ERG, PTEN and SPINK1 genomic alteration occur less frequently and the enrichment of ERG for PTEN loss is not observed. Additionally, patients with combined PTEN loss/ERG positive are at highest risk for BCR vs North American Caucasian population where PTEN loss alone seems to be associated with the worst clinical outcome. The data presented here further support differences in clonal evolution between Middle Eastern and Western population in relation to PCA and add further insight to understanding PCA molecular pathways.
Collapse
Affiliation(s)
- Ramy A Abdelsalam
- Department of Pathology and Laboratory Medicine, University of Calgary and Calgary Laboratory Services, Calgary, AB, Canada
- Department of Pathology, Mansoura University, Mansoura, Egypt
| | - Ibrahim Khalifeh
- Department of Pathology and Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Alan Box
- Department of Pathology and Laboratory Medicine, University of Calgary and Calgary Laboratory Services, Calgary, AB, Canada
| | - Maria Kalantarian
- Department of Pathology and Laboratory Medicine, University of Calgary and Calgary Laboratory Services, Calgary, AB, Canada
| | - Sunita Ghosh
- Alberta Health Services-Cancer Control and Department of Medical Oncology, University of Alberta, Edmonton, AB, Canada
| | - Hatem Abou-Ouf
- Department of Pathology and Laboratory Medicine, University of Calgary and Calgary Laboratory Services, Calgary, AB, Canada
| | - Tamara Lotfi
- Department of Pathology and Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Mohammed Shahait
- Department of Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Nallasivam Palanisamy
- Department of Urology, Vattikuti Urology Institute, Henry Ford Health System, Detroit, MI, USA
| | - Tarek A Bismar
- Department of Pathology and Laboratory Medicine, University of Calgary and Calgary Laboratory Services, Calgary, AB, Canada.
- Departments of Oncology, Biochemistry and Molecular Biology, Calgary, AB, Canada.
- Arnie Charbonneau Cancer Institute and Tom Baker Cancer Center, Calgary, AB, Canada.
- Rockyview General Hospital, 7007, 14th st, Calgary, SW, ABT2V1P9, Canada.
| |
Collapse
|
38
|
Li J, Zhang B, Liu M, Fu X, Ci X, A J, Fu C, Dong G, Wu R, Zhang Z, Fu L, Dong JT. KLF5 Is Crucial for Androgen-AR Signaling to Transactivate Genes and Promote Cell Proliferation in Prostate Cancer Cells. Cancers (Basel) 2020; 12:cancers12030748. [PMID: 32245249 PMCID: PMC7140031 DOI: 10.3390/cancers12030748] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/15/2020] [Accepted: 03/17/2020] [Indexed: 01/08/2023] Open
Abstract
Androgen/androgen receptor (AR) signaling drives both the normal prostate development and prostatic carcinogenesis, and patients with advanced prostate cancer often develop resistance to androgen deprivation therapy. The transcription factor Krüppel-like factor 5 (KLF5) also regulates both normal and cancerous development of the prostate. In this study, we tested whether and how KLF5 plays a role in the function of AR signaling in prostate cancer cells. We found that KLF5 is upregulated by androgen depending on AR in LNCaP and C4-2B cells. Silencing KLF5, in turn, reduced AR transcriptional activity and inhibited androgen-induced cell proliferation and tumor growth in vitro and in vivo. Mechanistically, KLF5 occupied the promoter of AR, and silencing KLF5 repressed AR transcription. In addition, KLF5 and AR physically interacted with each other to regulate the expression of multiple genes (e.g., MYC, CCND1 and PSA) to promote cell proliferation. These findings indicate that, while transcriptionally upregulated by AR signaling, KLF5 also regulates the expression and transcriptional activity of AR in androgen-sensitive prostate cancer cells. The KLF5-AR interaction could provide a therapeutic opportunity for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Juan Li
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China (L.F.)
- School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen, Guangdong 518055, China;
| | - Baotong Zhang
- Emory Winship Cancer Institute, Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365-C Clifton Road, Atlanta, GA 30322, USA
| | - Mingcheng Liu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China (L.F.)
- School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen, Guangdong 518055, China;
| | - Xing Fu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China (L.F.)
- School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen, Guangdong 518055, China;
| | - Xinpei Ci
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Jun A
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China (L.F.)
- School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen, Guangdong 518055, China;
| | - Changying Fu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China (L.F.)
- School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen, Guangdong 518055, China;
| | - Ge Dong
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China (L.F.)
| | - Rui Wu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China (L.F.)
- School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen, Guangdong 518055, China;
| | - Zhiqian Zhang
- School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen, Guangdong 518055, China;
| | - Liya Fu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China (L.F.)
| | - Jin-Tang Dong
- School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen, Guangdong 518055, China;
- Emory Winship Cancer Institute, Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365-C Clifton Road, Atlanta, GA 30322, USA
- Correspondence:
| |
Collapse
|
39
|
Luca BA, Moulton V, Ellis C, Edwards DR, Campbell C, Cooper RA, Clark J, Brewer DS, Cooper CS. A novel stratification framework for predicting outcome in patients with prostate cancer. Br J Cancer 2020; 122:1467-1476. [PMID: 32203215 PMCID: PMC7217762 DOI: 10.1038/s41416-020-0799-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 02/05/2020] [Accepted: 02/26/2020] [Indexed: 12/25/2022] Open
Abstract
Background Unsupervised learning methods, such as Hierarchical Cluster Analysis, are commonly used for the analysis of genomic platform data. Unfortunately, such approaches ignore the well-documented heterogeneous composition of prostate cancer samples. Our aim is to use more sophisticated analytical approaches to deconvolute the structure of prostate cancer transcriptome data, providing novel clinically actionable information for this disease. Methods We apply an unsupervised model called Latent Process Decomposition (LPD), which can handle heterogeneity within individual cancer samples, to genome-wide expression data from eight prostate cancer clinical series, including 1,785 malignant samples with the clinical endpoints of PSA failure and metastasis. Results We show that PSA failure is correlated with the level of an expression signature called DESNT (HR = 1.52, 95% CI = [1.36, 1.7], P = 9.0 × 10−14, Cox model), and that patients with a majority DESNT signature have an increased metastatic risk (X2 test, P = 0.0017, and P = 0.0019). In addition, we develop a stratification framework that incorporates DESNT and identifies three novel molecular subtypes of prostate cancer. Conclusions These results highlight the importance of using more complex approaches for the analysis of genomic data, may assist drug targeting, and have allowed the construction of a nomogram combining DESNT with other clinical factors for use in clinical management.
Collapse
Affiliation(s)
- Bogdan-Alexandru Luca
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, Norfolk, UK.,School of Computing Sciences, University of East Anglia, Norwich Research Park, Norwich, Norfolk, UK
| | - Vincent Moulton
- School of Computing Sciences, University of East Anglia, Norwich Research Park, Norwich, Norfolk, UK
| | - Christopher Ellis
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, Norfolk, UK.,School of Computing Sciences, University of East Anglia, Norwich Research Park, Norwich, Norfolk, UK
| | - Dylan R Edwards
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, Norfolk, UK
| | - Colin Campbell
- Intelligent Systems Laboratory, University of Bristol, Bristol, UK
| | - Rosalin A Cooper
- Department of Pathology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Jeremy Clark
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, Norfolk, UK
| | - Daniel S Brewer
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, Norfolk, UK.,The Earlham Institute, Norwich Research Park, Norwich, Norfolk, UK
| | - Colin S Cooper
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, Norfolk, UK.
| |
Collapse
|
40
|
The Genomic and Molecular Pathology of Prostate Cancer: Clinical Implications for Diagnosis, Prognosis, and Therapy. Adv Anat Pathol 2020; 27:11-19. [PMID: 31503032 DOI: 10.1097/pap.0000000000000245] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Prostate cancer (PCa) is the most common noncutaneous malignancy affecting American men and the second most common cause of cancer death. The traditional risk classification schemes for PCa are limited due to the vast clinical and molecular heterogeneity of the disease. Fortunately, recent advancements in sequencing technologies have provided us with valuable insight into the genomics of PCa. To date, a wide array of recurrent genomic alterations in PCa have been identified. Incorporating these distinct molecular subtypes of PCa into prediction models provides opportunities for improved risk stratification and ultimately better patient outcomes. In this review, we summarize the key molecular subtypes of PCa and focus on those genomic alterations that have clinical implications for diagnosis, prognosis, and therapeutic response.
Collapse
|
41
|
Shang Z, Yu J, Sun L, Tian J, Zhu S, Zhang B, Dong Q, Jiang N, Flores-Morales A, Chang C, Niu Y. LncRNA PCAT1 activates AKT and NF-κB signaling in castration-resistant prostate cancer by regulating the PHLPP/FKBP51/IKKα complex. Nucleic Acids Res 2019; 47:4211-4225. [PMID: 30773595 PMCID: PMC6486551 DOI: 10.1093/nar/gkz108] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 01/14/2019] [Accepted: 02/12/2019] [Indexed: 12/14/2022] Open
Abstract
In PTEN-deficient prostate cancers, AKT signaling may be activated upon suppression of androgen receptor signaling. Activation of AKT as well as NF-κB signaling involves a key regulatory protein complex containing PHLPP, FKBP51 and IKKα. Here, we report a critical role of lncRNA PCAT1 in regulating the PHLPP/FKBP51/IKKα complex and progression of castration-resistant prostate cancer (CRPC). Using database queries, bioinformatic analyses, as well as RIP and RNA pull-down assays, we discovered and validated that the lncRNA-PCAT1 perturbs the PHLPP/FKBP51/IKKα complex and activates AKT and NF-κB signaling. Expression of lncRNA-PCAT1 is positively linked to CRPC progression. PCAT1 binds directly to FKBP51, displacing PHLPP from the PHLPP/FKBP51/IKKα complex, leading to activation of AKT and NF-κB signaling. Targeting PCAT1 restores PHLPP binding to FKBP1 leading to suppression of AKT signaling. Preclinical study in a mouse model of CRPC suggests therapeutic potential by targeting lncRNA PCAT1 to suppress CRPC progression. Together, the newly identified PCAT1/FKBP51/IKKα complex provides mechanistic insight in the interplay between AKT, NF-κB and AR signaling in CRPC, and the preclinical studies suggest that a novel role for PCAT1 as a therapeutic target.
Collapse
Affiliation(s)
- Zhiqun Shang
- Tianjin Institute of Urology, the 2nd Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Jianpeng Yu
- Tianjin Institute of Urology, the 2nd Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Libin Sun
- Tianjin Institute of Urology, the 2nd Hospital of Tianjin Medical University, Tianjin 300211, China.,Department of Urology, First Affiliated Hospital, Shanxi Medical University, Shanxi 030001, China
| | - Jing Tian
- Tianjin Institute of Urology, the 2nd Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Shimiao Zhu
- Tianjin Institute of Urology, the 2nd Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Boya Zhang
- Tianjin Institute of Urology, the 2nd Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Qian Dong
- Tianjin Institute of Urology, the 2nd Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Ning Jiang
- Tianjin Institute of Urology, the 2nd Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Amilcar Flores-Morales
- Department of Health Science, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Chawnshang Chang
- Tianjin Institute of Urology, the 2nd Hospital of Tianjin Medical University, Tianjin 300211, China.,Department of Pathology and Urology, University of Rochester, Rochester, NY 14620, USA
| | - Yuanjie Niu
- Tianjin Institute of Urology, the 2nd Hospital of Tianjin Medical University, Tianjin 300211, China
| |
Collapse
|
42
|
DEPTOR is an in vivo tumor suppressor that inhibits prostate tumorigenesis via the inactivation of mTORC1/2 signals. Oncogene 2019; 39:1557-1571. [PMID: 31685947 PMCID: PMC7018663 DOI: 10.1038/s41388-019-1085-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 01/06/2023]
Abstract
The DEPTOR-mTORC1/2 axis has been shown to play an important, but a context dependent role in the regulation of proliferation and the survival of various cancer cells in cell culture settings. The in vivo role of DEPTOR in tumorigenesis remains elusive. Here we showed that the levels of both DEPTOR protein and mRNA were substantially decreased in human prostate cancer tissues, which positively correlated with disease progression. DEPTOR depletion accelerated proliferation and survival, migration, and invasion in human prostate cancer cells. Mechanistically, DEPTOR depletion not only activated both mTORC1 and mTORC2 signals to promote cell proliferation and survival, but also induced an AKT-dependent epithelial–mesenchymal transition (EMT) and β-catenin nuclear translocation to promote cell migration and invasion. Abrogation of mTOR or AKT activation rescued the biological consequences of DEPTOR depletion. Importantly, in a Deptor-KO mouse model, Deptor knockout accelerated prostate tumorigenesis triggered by Pten loss via the activation of mTOR signaling. Collectively, our study demonstrates that DEPTOR is a tumor suppressor in the prostate, and its depletion promotes tumorigenesis via the activation of mTORC1 and mTORC2 signals. Thus, DEPTOR reactivation via a variety of means would have therapeutic potential for the treatment of prostate cancer.
Collapse
|
43
|
Hasanzad M, Sarhangi N, Aghaei Meybodi HR, Nikfar S, Khatami F, Larijani B. Precision Medicine in Non Communicable Diseases. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2019; 8:1-18. [PMID: 32351905 PMCID: PMC7175610 DOI: 10.22088/ijmcm.bums.8.2.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 07/20/2019] [Indexed: 12/12/2022]
Abstract
Non-communicable diseases (NCDs) are the leading cause of death and disease burden globally, cardiovascular diseases (CVDs) account for the major part of death related to NCDs followed by different types of cancer, chronic obstructive pulmonary disease (COPD), and diabetes. As the World Health Organization (WHO) and the United Nations have announced a 25% reduction in mortality of NCDs by 2025, different communities need to adopt preventive strategies for achieving this goal. Personalized medicine approach as a predictive and preventive strategy aims for a better therapeutic goal to the patients to maximize benefits and reduce harms. The clinical benefits of this approach are already realized in cancer targeted therapy, and its impact on other conditions needs more studies in different societies. In this review, we essentially describe the concept of personalized (or precision) medicine in association with NCDs and the future of precision medicine in prediction, prevention, and personalized treatment.
Collapse
Affiliation(s)
- Mandana Hasanzad
- Medical Genomics Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.,Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Negar Sarhangi
- Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Aghaei Meybodi
- Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shekoufeh Nikfar
- Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pharmacoeconomics and Pharmaceutical Administration, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Khatami
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.,Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
44
|
Abou-Ouf H, Ghosh S, Box A, Palanisamy N, Bismar TA. Combined loss of TFF3 and PTEN is associated with lethal outcome and overall survival in men with prostate cancer. J Cancer Res Clin Oncol 2019; 145:1751-1759. [PMID: 31129769 DOI: 10.1007/s00432-019-02933-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 05/08/2019] [Indexed: 01/03/2023]
Abstract
BACKGROUND Trefoil Factor 3 (TFF3) has been implicated in Prostate Cancer (PCa) progression. However, its prognostic value and association with other biomarkers have not been fully explored. We assessed the combined value of TFF3 and PTEN in two cohorts: one is managed surgically for localized PCa and the second is managed non-surgically by androgen deprivation therapy for advanced disease. DESIGN 228 radical prostatectomies (RP) and 318 transurethral resection of prostate (TURP) samples were assessed by immunohistochemistry (IHC) for TFF3 and by IHC and fluorescent in situ hybridization (FISH) for PTEN. Results of biomarkers expression were correlated with various pathological and clinical outcome parameters including biochemical recurrence (BCR) in the RP cohort and cancer-specific mortality (PCSM) and overall survival (OS) in the TURP cohort. RESULTS TFF3 expression was detected in 131/226 (57.9%) RP samples and 148/318 (46.5%) of TURP cases. In general, TFF3 positivity was less frequently observed with advanced Gleason Groups. TFF3 expression was also assessed in relation to PTEN expression. Only 15-16% of TFF3-expressed cases were present in association with complete loss of PTEN expression in the TURP and localized cohorts, respectively. Loss of TFF3 expression was not related to BCR after RP, but was prognostic in the non-surgical cohort and associated with decrease OS and PCSM (HR 2.31, CI: 1.67-3.18, p < 0.0001) and (HR 3.99, CI: 2.43-6.56; p < 0.0001), respectively. Adjusting for Gleason score, combined loss of TFF3/PTEN was most associated with OS (HR 2.33, CI: 1.49-3.62; p < 0.0001) and PCSM (HR = 3.44, CI: 1.75-6.78, p < 0.0001). CONCLUSION The study documents for the first time significant association for combined status of TFF3 expression and PTEN loss in OS and PCSM in patients not managed by surgical intervention. Prospective assessment of PTEN and TFF3 may provide further insight into molecularly subtyping PCa and aid in stratifying patients at risk for lethal disease.
Collapse
Affiliation(s)
- Hatem Abou-Ouf
- Department of Pathology and Laboratory Medicine, University of Calgary Cumming School of Medicine and Calgary Laboratory Services, Calgary, AB, Canada
| | - Sunita Ghosh
- Department of Medical Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Adrian Box
- Department of Pathology and Laboratory Medicine, University of Calgary Cumming School of Medicine and Calgary Laboratory Services, Calgary, AB, Canada
| | - Nallasivam Palanisamy
- Department of Urology, Vattikuti Urology Institute, Henry Ford Health System Detroit, Detroit, MI, USA
| | - Tarek A Bismar
- Department of Pathology and Laboratory Medicine, University of Calgary Cumming School of Medicine and Calgary Laboratory Services, Calgary, AB, Canada.
- Department of Oncology, University of Calgary Cumming School of Medicine, Calgary, AB, Canada.
- Department of Biochemistry and Molecular Biology, University of Calgary Cumming School of Medicine, Calgary, AB, Canada.
- Arnie Charbonneau Cancer Institute and Tom Baker Cancer Center, Calgary, AB, Canada.
| |
Collapse
|
45
|
Khan MI, Al Johani A, Hamid A, Ateeq B, Manzar N, Adhami VM, Lall RK, Rath S, Sechi M, Siddiqui IA, Choudhry H, Zamzami MA, Havighurst TC, Huang W, Ntambi JM, Mukhtar H. Proproliferative function of adaptor protein GRB10 in prostate carcinoma. FASEB J 2019; 33:3198-3211. [PMID: 30379590 PMCID: PMC6404554 DOI: 10.1096/fj.201800265rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Growth factor receptor-binding protein 10 (GRB10) is a well-known adaptor protein and a recently identified substrate of the mammalian target of rapamycin (mTOR). Depletion of GRB10 increases insulin sensitivity and overexpression suppresses PI3K/Akt signaling. Because the major reason for the limited efficacy of PI3K/Akt-targeted therapies in prostate cancer (PCa) is loss of mTOR-regulated feedback suppression, it is therefore important to assess the functional importance and regulation of GRB10 under these conditions. On the basis of these background observations, we explored the status and functional impact of GRB10 in PCa and found maximum expression in phosphatase and tensin homolog (PTEN)-deficient PCa. In human PCa samples, GRB10 inversely correlated with PTEN and positively correlated with pAKT levels. Knockdown of GRB10 in nontumorigenic PTEN null mouse embryonic fibroblasts and tumorigenic PCa cell lines reduced Akt phosphorylation and selectively activated a panel of receptor tyrosine kinases. Similarly, overexpression of GRB10 in PTEN wild-type PCa cell lines accelerated tumorigenesis and induced Akt phosphorylation. In PTEN wild-type PCa, GRB10 overexpression promoted mediated PTEN interaction and degradation. PI3K (but not mTOR) inhibitors reduced GRB10 expression, suggesting primarily PI3K-driven regulation of GRB10. In summary, our results suggest that GRB10 acts as a major downstream effector of PI3K and has tumor-promoting effects in prostate cancer.-Khan, M. I., Al Johani, A., Hamid, A., Ateeq, B., Manzar, N., Adhami, V. M., Lall, R. K., Rath, S., Sechi, M., Siddiqui, I. A., Choudhry, H., Zamzami, M. A., Havighurst, T. C., Huang, W., Ntambi, J. M., Mukhtar, H. Proproliferatve function of adaptor protein GRB10 in prostate carcinoma.
Collapse
Affiliation(s)
- Mohammad Imran Khan
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia;,Cancer Metabolism and Epigenetic Unit, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia;,Department of Dermatology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA;,Correspondence: Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia. E-mail:
| | - Ahmed Al Johani
- Department of Biochemistry, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | - Abid Hamid
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | - Bushra Ateeq
- Department of Biological Sciences and Bioengineering, Molecular Oncology Laboratory, Indian Institute of Technology–Kanpur (IIT–K), Kanpur, India
| | - Nishat Manzar
- Department of Biological Sciences and Bioengineering, Molecular Oncology Laboratory, Indian Institute of Technology–Kanpur (IIT–K), Kanpur, India
| | - Vaqar Mustafa Adhami
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | - Rahul K. Lall
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | - Suvasmita Rath
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | - Mario Sechi
- Department of Chemistry and Pharmacy, University of Sassari, Sassari, Italy
| | - Imtiaz Ahmad Siddiqui
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | - Hani Choudhry
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia;,Cancer Metabolism and Epigenetic Unit, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mazin A. Zamzami
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia;,Cancer Metabolism and Epigenetic Unit, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Thomas C. Havighurst
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | - Wei Huang
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | - James M. Ntambi
- Department of Biochemistry, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | - Hasan Mukhtar
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| |
Collapse
|
46
|
Tosoian JJ, Guedes LB, Morais CL, Mamawala M, Ross AE, De Marzo AM, Trock BJ, Han M, Carter HB, Lotan TL. PTEN status assessment in the Johns Hopkins active surveillance cohort. Prostate Cancer Prostatic Dis 2019; 22:176-181. [PMID: 30279579 PMCID: PMC6372343 DOI: 10.1038/s41391-018-0093-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 07/10/2018] [Accepted: 07/20/2018] [Indexed: 01/03/2023]
Abstract
BACKGROUND Up to half of men with Gleason score 6 (GS6) prostate cancers initially managed with active surveillance (AS) will eventually require definitive therapy, usually due to tumor grade reclassification during follow-up. We examined the association between PTEN status on biopsy and subsequent clinicopathologic outcomes in men with GS6 cancers who enrolled in AS. METHODS We performed a case-control study of men enrolled in the Johns Hopkins AS cohort with diagnostic biopsy tissue available for immunohistochemical (IHC) staining. IHC was performed for PTEN using genetically validated protocols for all patients. Cases included men who underwent grade reclassification to GS ≥ 3 + 4 = 7 on biopsy within 2 years of follow-up (i.e., early reclassification) or reclassification to GS ≥ 4 + 3 = 7 on biopsy or radical prostatectomy during follow-up (i.e., extreme reclassification). Control patients were diagnosed with GS6 cancer and monitored on AS for at least 8 years without undergoing biopsy reclassification. RESULTS Among 67 cases with adequate tissue, 31 men underwent early reclassification and 36 men underwent extreme reclassification. Cases were compared to 65 control patients with adequate tissue for assessment. On initial prostate biopsy, cases were older (median age 67 vs. 65, p = 0.024) and were less likely to meet very-low-risk criteria (64 vs 79%, p = 0.042) as compared to controls. Although not statistically significant, PTEN loss was observed in only 1 (1.5%) of 65 controls as compared to 6 (9%) of 67 cases (p = 0.062). CONCLUSIONS PTEN loss was rare among men with GS6 prostate cancer enrolled in AS at Johns Hopkins. Despite this, PTEN loss was more frequent among men who underwent early or extreme reclassification to higher-grade cancer as compared to controls. Additional studies in larger low-risk cohorts may better elucidate a potential role for PTEN in selecting patients for AS.
Collapse
Affiliation(s)
- Jeffrey J Tosoian
- Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Liana B Guedes
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Carlos L Morais
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Mufaddal Mamawala
- Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ashley E Ross
- Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Angelo M De Marzo
- Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Bruce J Trock
- Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Misop Han
- Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | - Tamara L Lotan
- Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
47
|
Tian T, Li X, Zhang J. mTOR Signaling in Cancer and mTOR Inhibitors in Solid Tumor Targeting Therapy. Int J Mol Sci 2019; 20:ijms20030755. [PMID: 30754640 PMCID: PMC6387042 DOI: 10.3390/ijms20030755] [Citation(s) in RCA: 403] [Impact Index Per Article: 67.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 01/28/2019] [Accepted: 02/01/2019] [Indexed: 12/12/2022] Open
Abstract
The mammalian or mechanistic target of rapamycin (mTOR) pathway plays a crucial role in regulation of cell survival, metabolism, growth and protein synthesis in response to upstream signals in both normal physiological and pathological conditions, especially in cancer. Aberrant mTOR signaling resulting from genetic alterations from different levels of the signal cascade is commonly observed in various types of cancers. Upon hyperactivation, mTOR signaling promotes cell proliferation and metabolism that contribute to tumor initiation and progression. In addition, mTOR also negatively regulates autophagy via different ways. We discuss mTOR signaling and its key upstream and downstream factors, the specific genetic changes in the mTOR pathway and the inhibitors of mTOR applied as therapeutic strategies in eight solid tumors. Although monotherapy and combination therapy with mTOR inhibitors have been extensively applied in preclinical and clinical trials in various cancer types, innovative therapies with better efficacy and less drug resistance are still in great need, and new biomarkers and deep sequencing technologies will facilitate these mTOR targeting drugs benefit the cancer patients in personalized therapy.
Collapse
Affiliation(s)
- Tian Tian
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China.
| | - Xiaoyi Li
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China.
| | - Jinhua Zhang
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China.
| |
Collapse
|
48
|
Hernández G, Ramírez JL, Pedroza-Torres A, Herrera LA, Jiménez-Ríos MA. The Secret Life of Translation Initiation in Prostate Cancer. Front Genet 2019; 10:14. [PMID: 30761182 PMCID: PMC6363655 DOI: 10.3389/fgene.2019.00014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 01/11/2019] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is the second most prevalent cancer in men worldwide. Despite the advances understanding the molecular processes driving the onset and progression of this disease, as well as the continued implementation of screening programs, PCa still remains a significant cause of morbidity and mortality, in particular in low-income countries. It is only recently that defects of the translation process, i.e., the synthesis of proteins by the ribosome using a messenger (m)RNA as a template, have begun to gain attention as an important cause of cancer development in different human tissues, including prostate. In particular, the initiation step of translation has been established to play a key role in tumorigenesis. In this review, we discuss the state-of-the-art of three key aspects of protein synthesis in PCa, namely, misexpression of translation initiation factors, dysregulation of the major signaling cascades regulating translation, and the therapeutic strategies based on pharmacological compounds targeting translation as a novel alternative to those based on hormones controlling the androgen receptor pathway.
Collapse
Affiliation(s)
- Greco Hernández
- Translation and Cancer Laboratory, Unit of Biomedical Research on Cancer, National Institute of Cancer, Mexico City, Mexico
| | - Jorge L. Ramírez
- Translation and Cancer Laboratory, Unit of Biomedical Research on Cancer, National Institute of Cancer, Mexico City, Mexico
| | - Abraham Pedroza-Torres
- Cátedra-CONACyT Program, Hereditary Cancer Clinic, National Institute of Cancer, Mexico City, Mexico
| | - Luis A. Herrera
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, The National Autonomous University of Mexico, Mexico City, Mexico
| | | |
Collapse
|
49
|
Batra A, Winquist E. Emerging cell cycle inhibitors for treating metastatic castration-resistant prostate cancer. Expert Opin Emerg Drugs 2018; 23:271-282. [PMID: 30422005 DOI: 10.1080/14728214.2018.1547707] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Disease progression despite androgen suppression defines lethal castration-resistant prostate cancer (CRPC). Most of these cancers remain androgen receptor (AR)-signaling dependent. Therapy for metastatic CRPC includes abiraterone acetate, enzalutamide, docetaxel, cabazitaxel, sipuleucel-T, and radium-223. However, survival remains modest for men with progressive disease despite AR-targeted therapy and docetaxel, and therefore novel treatments are needed. Areas covered: Recent evidence of genomic heterogeneity and sensitivity to PARP inhibitors supports investigation of targeted agents in CRPC. Cell cycle inhibitors are therefore logical molecules to investigate. Review of the current literature identified cell cycle inhibitors under study in early phase clinical trials targeting the G1 (palbociclib, ribociclib, AZD-5363, ipatasertib), S (M-6620, prexasertib), G2 (adavosertib), and M (alisertib) phases of the cell cycle. Expert opinion: Strategies combining cell cycle inhibitors with active agents in CRPC are most likely to have clinical impact with CDK4/6 and Wee1 inhibitors appearing most promising. Identification of predictive biomarkers may be essential and currently trials are testing circulating cell-free DNA as an approach. Incremental toxicities such as neutropenia are important in this population. Results from most current clinical trials of cell cycle inhibitors in CRPC are still pending but it is anticipated they will provide important insights into the heterogeneous biology of CRPC.
Collapse
Affiliation(s)
- Anupam Batra
- a Division of Medical Oncology, Department of Oncology, Schulich School of Medicine & Dentistry , Western University and London Health Sciences Centre , London , ON , Canada
| | - Eric Winquist
- a Division of Medical Oncology, Department of Oncology, Schulich School of Medicine & Dentistry , Western University and London Health Sciences Centre , London , ON , Canada
| |
Collapse
|
50
|
Iczkowski KA. Large-Gland Proliferations of the Prostate. Surg Pathol Clin 2018; 11:687-712. [PMID: 30447836 DOI: 10.1016/j.path.2018.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Large-gland proliferations of the prostate have gained considerable attention in the past decade. The differential diagnosis is quite broad but can be refined using histologic criteria and, sometimes, immunostains. Pathologists have come to realize that cribriform and intraductal as well as ductal carcinomas are particularly aggressive patterns, and should name them in diagnostic reporting when present.
Collapse
Affiliation(s)
- Kenneth A Iczkowski
- Department of Pathology, Medical College of Wisconsin, 9200 West Wisconsin Avenue, Milwaukee, WI 53226, USA.
| |
Collapse
|