1
|
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
|
2
|
Al Bashir S, Alorjani MS, Kheirallah K, Al Hamad M, Haddad HK, Al-Dwairy A, Bani-Fawwaz BA, Aldaoud N, Halalsheh O, Amawi S, Matalka II. PTEN, ERG, SPINK1, and TFF3 Status and Relationship in a Prostate Cancer Cohort from Jordanian Arab Population. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:174. [PMID: 38256434 PMCID: PMC10821453 DOI: 10.3390/medicina60010174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/11/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024]
Abstract
Background and Objectives: Prognostic biomarkers in prostate cancer (PCa) include PTEN, ERG, SPINK1, and TFF3. Their relationships and patterns of expression in PCa in developing countries, including Jordan, have not yet been investigated. Materials and Methods: A tissue microarray (TMA) of PCa patients was taken from paraffin-embedded tissue blocks for 130 patients. PTEN, ERG, SPINK1, and TFF3 expression profiles were examined using immunohistochemistry (IHC) and correlated with each other and other clinicopathological factors. Results: PTEN loss of any degree was observed in 42.9% of PCa cases. ERG and TFF3 were expressed in 59.3% and 46.5% of PCa cases, respectively. SPINK1 expression was observed in 6 out of 104 PCa cases (5.4%). Among all PCa cases (n = 104), 3.8% (n = 4) showed SPINK1+/ERG+ phenotype, 1.9% (n = 2) showed SPINK1+/ERG- phenotype, 56.7% (n = 59) showed SPINK1-/ERG+ phenotype, and 37.5% showed SPINK1-/ERG- phenotype (n = 39). Among ERG positive cases (n = 63), 6.3% were SPINK1 positive. Among SPINK1 positive cases (n = 6), 66.7% were ERG positive. SPINK1 expression was predominantly observed in a subgroup of cancers that expressed TFF3 (6/6). Additionally, a statistically significant loss of PTEN expression was observed from Gleason Score 6 (GS6) (Grade Group 1 (GG1)) to GS9-10 (GG5); (p-value 0.019). Conclusions: This is the first study to look at the status of the PTEN, ERG, SPINK1, and TFF3 genes in a Jordanian Arab population. Loss of PTEN has been linked to more aggressive prostate cancer with high GSs/GGs. SPINK1 expression was predominantly observed in a subgroup of cancers that expressed TFF3. Our results call for screening these biomarkers for grading and molecular subtyping of the disease.
Collapse
Affiliation(s)
- Samir Al Bashir
- Department of Pathology and Microbiology, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan; (M.S.A.); (N.A.); (I.I.M.)
| | - Mohammed S. Alorjani
- Department of Pathology and Microbiology, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan; (M.S.A.); (N.A.); (I.I.M.)
| | - Khalid Kheirallah
- Department of Public Health and Community Medicine, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan;
| | - Mohammad Al Hamad
- Department of Pathology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia;
| | - Husam K. Haddad
- Department of Pathology and Laboratory Medicine, Ministry of Health, Amman 11118, Jordan;
| | - Ahmad Al-Dwairy
- Medstar-Georgetown Washington Hospital Center, Georgetown University, Washington, DC 20057, USA;
| | - Baha A. Bani-Fawwaz
- Gastroenterology and Hepatology Department, Adventhealth, Orlando, FL 32804, USA;
| | - Najla Aldaoud
- Department of Pathology and Microbiology, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan; (M.S.A.); (N.A.); (I.I.M.)
| | - Omar Halalsheh
- Department of General Surgery and Urology, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan;
| | - Saddam Amawi
- Johns Hopkins Aramco Health Centre, Al Mubarraz 36423, Saudi Arabia;
| | - Ismail I. Matalka
- Department of Pathology and Microbiology, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan; (M.S.A.); (N.A.); (I.I.M.)
- College of Medicine, Ras Al-Khaimah (RAK) Medical and Health Sciences University, Ras Al-Khaimah 11172, United Arab Emirates
| |
Collapse
|
3
|
Travis G, McGowan EM, Simpson AM, Marsh DJ, Nassif NT. PTEN, PTENP1, microRNAs, and ceRNA Networks: Precision Targeting in Cancer Therapeutics. Cancers (Basel) 2023; 15:4954. [PMID: 37894321 PMCID: PMC10605164 DOI: 10.3390/cancers15204954] [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: 09/11/2023] [Revised: 10/06/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
The phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is a well characterised tumour suppressor, playing a critical role in the maintenance of fundamental cellular processes including cell proliferation, migration, metabolism, and survival. Subtle decreases in cellular levels of PTEN result in the development and progression of cancer, hence there is tight regulation of the expression, activity, and cellular half-life of PTEN at the transcriptional, post-transcriptional, and post-translational levels. PTENP1, the processed pseudogene of PTEN, is an important transcriptional and post-transcriptional regulator of PTEN. PTENP1 expression produces sense and antisense transcripts modulating PTEN expression, in conjunction with miRNAs. Due to the high sequence similarity between PTEN and the PTENP1 sense transcript, the transcripts possess common miRNA binding sites with the potential for PTENP1 to compete for the binding, or 'sponging', of miRNAs that would otherwise target the PTEN transcript. PTENP1 therefore acts as a competitive endogenous RNA (ceRNA), competing with PTEN for the binding of specific miRNAs to alter the abundance of PTEN. Transcription from the antisense strand produces two functionally independent isoforms (PTENP1-AS-α and PTENP1-AS-β), which can regulate PTEN transcription. In this review, we provide an overview of the post-transcriptional regulation of PTEN through interaction with its pseudogene, the cellular miRNA milieu and operation of the ceRNA network. Furthermore, its importance in maintaining cellular integrity and how disruption of this PTEN-miRNA-PTENP1 axis may lead to cancer but also provide novel therapeutic opportunities, is discussed. Precision targeting of PTENP1-miRNA mediated regulation of PTEN may present as a viable alternative therapy.
Collapse
Affiliation(s)
- Glena Travis
- Cancer Biology, Faculty of Science, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; (G.T.); (E.M.M.)
| | - Eileen M. McGowan
- Cancer Biology, Faculty of Science, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; (G.T.); (E.M.M.)
- Central Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Ann M. Simpson
- Gene Therapy and Translational Molecular Analysis Laboratory, Faculty of Science, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia;
| | - Deborah J. Marsh
- Translational Oncology Group, Faculty of Science, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia;
| | - Najah T. Nassif
- Cancer Biology, Faculty of Science, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; (G.T.); (E.M.M.)
| |
Collapse
|
4
|
Logotheti S, Papadaki E, Zolota V, Logothetis C, Vrahatis AG, Soundararajan R, Tzelepi V. Lineage Plasticity and Stemness Phenotypes in Prostate Cancer: Harnessing the Power of Integrated "Omics" Approaches to Explore Measurable Metrics. Cancers (Basel) 2023; 15:4357. [PMID: 37686633 PMCID: PMC10486655 DOI: 10.3390/cancers15174357] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/21/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Prostate cancer (PCa), the most frequent and second most lethal cancer type in men in developed countries, is a highly heterogeneous disease. PCa heterogeneity, therapy resistance, stemness, and lethal progression have been attributed to lineage plasticity, which refers to the ability of neoplastic cells to undergo phenotypic changes under microenvironmental pressures by switching between developmental cell states. What remains to be elucidated is how to identify measurements of lineage plasticity, how to implement them to inform preclinical and clinical research, and, further, how to classify patients and inform therapeutic strategies in the clinic. Recent research has highlighted the crucial role of next-generation sequencing technologies in identifying potential biomarkers associated with lineage plasticity. Here, we review the genomic, transcriptomic, and epigenetic events that have been described in PCa and highlight those with significance for lineage plasticity. We further focus on their relevance in PCa research and their benefits in PCa patient classification. Finally, we explore ways in which bioinformatic analyses can be used to determine lineage plasticity based on large omics analyses and algorithms that can shed light on upstream and downstream events. Most importantly, an integrated multiomics approach may soon allow for the identification of a lineage plasticity signature, which would revolutionize the molecular classification of PCa patients.
Collapse
Affiliation(s)
- Souzana Logotheti
- Department of Pathology, University of Patras, 26504 Patras, Greece; (S.L.); (E.P.); (V.Z.)
| | - Eugenia Papadaki
- Department of Pathology, University of Patras, 26504 Patras, Greece; (S.L.); (E.P.); (V.Z.)
- Department of Informatics, Ionian University, 49100 Corfu, Greece;
| | - Vasiliki Zolota
- Department of Pathology, University of Patras, 26504 Patras, Greece; (S.L.); (E.P.); (V.Z.)
| | - Christopher Logothetis
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | | | - Rama Soundararajan
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Vasiliki Tzelepi
- Department of Pathology, University of Patras, 26504 Patras, Greece; (S.L.); (E.P.); (V.Z.)
| |
Collapse
|
5
|
Diop MK, Molina OE, Birlea M, LaRue H, Hovington H, Têtu B, Lacombe L, Bergeron A, Fradet Y, Trudel D. Leukocytic Infiltration of Intraductal Carcinoma of the Prostate: An Exploratory Study. Cancers (Basel) 2023; 15:cancers15082217. [PMID: 37190147 DOI: 10.3390/cancers15082217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/03/2023] [Accepted: 04/07/2023] [Indexed: 05/17/2023] Open
Abstract
Intraductal carcinoma of the prostate (IDC-P) is an aggressive histological subtype of prostate cancer (PCa) detected in approximately 20% of radical prostatectomy (RP) specimens. As IDC-P has been associated with PCa-related death and poor responses to standard treatment, the purpose of this study was to explore the immune infiltrate of IDC-P. Hematoxylin- and eosin-stained slides from 96 patients with locally advanced PCa who underwent RP were reviewed to identify IDC-P. Immunohistochemical staining of CD3, CD8, CD45RO, FoxP3, CD68, CD163, CD209 and CD83 was performed. For each slide, the number of positive cells per mm2 in the benign tissues, tumor margins, cancer and IDC-P was calculated. Consequently, IDC-P was found in a total of 33 patients (34%). Overall, the immune infiltrate was similar in the IDC-P-positive and the IDC-P-negative patients. However, FoxP3+ regulatory T cells (p < 0.001), CD68+ and CD163+ macrophages (p < 0.001 for both) and CD209+ and CD83+ dendritic cells (p = 0.002 and p = 0.013, respectively) were less abundant in the IDC-P tissues compared to the adjacent PCa. Moreover, the patients were classified as having immunologically "cold" or "hot" IDC-P, according to the immune-cell densities averaged in the total IDC-P or in the immune hotspots. The CD68/CD163/CD209-immune hotspots predicted metastatic dissemination (p = 0.014) and PCa-related death (p = 0.009) in a Kaplan-Meier survival analysis. Further studies on larger cohorts are necessary to evaluate the clinical utility of assessing the immune infiltrate of IDC-P with regards to patient prognosis and the use of immunotherapy for lethal PCa.
Collapse
Affiliation(s)
- Mame-Kany Diop
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (axe Cancer) and Institut du Cancer de Montréal, 900 Saint-Denis, Montréal, QC H2X 0A9, Canada
- Department of Pathology and Cellular Biology, Université de Montréal, 2900 Boulevard Édouard-Montpetit, Montréal, QC H3T 1J4, Canada
| | - Oscar Eduardo Molina
- Centre de Recherche du CHU de Québec-Université Laval (axe Oncologie), Hôpital L'Hôtel-Dieu de Québec, 9 McMahon, Québec, QC G1R 3S3, Canada
| | - Mirela Birlea
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (axe Cancer) and Institut du Cancer de Montréal, 900 Saint-Denis, Montréal, QC H2X 0A9, Canada
| | - Hélène LaRue
- Centre de Recherche du CHU de Québec-Université Laval (axe Oncologie), Hôpital L'Hôtel-Dieu de Québec, 9 McMahon, Québec, QC G1R 3S3, Canada
| | - Hélène Hovington
- Centre de Recherche du CHU de Québec-Université Laval (axe Oncologie), Hôpital L'Hôtel-Dieu de Québec, 9 McMahon, Québec, QC G1R 3S3, Canada
| | - Bernard Têtu
- Centre de Recherche du CHU de Québec-Université Laval (axe Oncologie), Hôpital L'Hôtel-Dieu de Québec, 9 McMahon, Québec, QC G1R 3S3, Canada
- Department of Pathology, CHU de Québec-Université Laval, 11 Côte du Palais, Québec, QC G1R 2J6, Canada
| | - Louis Lacombe
- Centre de Recherche du CHU de Québec-Université Laval (axe Oncologie), Hôpital L'Hôtel-Dieu de Québec, 9 McMahon, Québec, QC G1R 3S3, Canada
- Department of Surgery, Université Laval, 2325 rue de l'Université, Québec, QC G1V 0A6, Canada
| | - Alain Bergeron
- Centre de Recherche du CHU de Québec-Université Laval (axe Oncologie), Hôpital L'Hôtel-Dieu de Québec, 9 McMahon, Québec, QC G1R 3S3, Canada
- Department of Surgery, Université Laval, 2325 rue de l'Université, Québec, QC G1V 0A6, Canada
| | - Yves Fradet
- Centre de Recherche du CHU de Québec-Université Laval (axe Oncologie), Hôpital L'Hôtel-Dieu de Québec, 9 McMahon, Québec, QC G1R 3S3, Canada
- Department of Surgery, Université Laval, 2325 rue de l'Université, Québec, QC G1V 0A6, Canada
| | - Dominique Trudel
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (axe Cancer) and Institut du Cancer de Montréal, 900 Saint-Denis, Montréal, QC H2X 0A9, Canada
- Department of Pathology and Cellular Biology, Université de Montréal, 2900 Boulevard Édouard-Montpetit, Montréal, QC H3T 1J4, Canada
- Department of Pathology, Centre Hospitalier de l'Université de Montréal, 1051 Sanguinet, Montréal, QC H2X 0C1, Canada
| |
Collapse
|
6
|
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
|
7
|
PTEN Protein Phosphatase Activity Is Not Required for Tumour Suppression in the Mouse Prostate. Biomolecules 2022; 12:biom12101511. [PMID: 36291720 PMCID: PMC9599176 DOI: 10.3390/biom12101511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [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
|
8
|
Patierno BM, Foo WC, Allen T, Somarelli JA, Ware KE, Gupta S, Wise S, Wise JP, Qin X, Zhang D, Xu L, Li Y, Chen X, Inman BA, McCall SJ, Huang J, Kittles RA, Owzar K, Gregory S, Armstrong AJ, George DJ, Patierno SR, Hsu DS, Freedman JA. Characterization of a castrate-resistant prostate cancer xenograft derived from a patient of West African ancestry. Prostate Cancer Prostatic Dis 2022; 25:513-523. [PMID: 34645983 PMCID: PMC9005588 DOI: 10.1038/s41391-021-00460-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/31/2021] [Accepted: 09/15/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND Prostate cancer is a clinically and molecularly heterogeneous disease, with highest incidence and mortality among men of African ancestry. To date, prostate cancer patient-derived xenograft (PCPDX) models to study this disease have been difficult to establish because of limited specimen availability and poor uptake rates in immunodeficient mice. Ancestrally diverse PCPDXs are even more rare, and only six PCPDXs from self-identified African American patients from one institution were recently made available. METHODS In the present study, we established a PCPDX from prostate cancer tissue from a patient of estimated 90% West African ancestry with metastatic castration resistant disease, and characterized this model's pathology, karyotype, hotspot mutations, copy number, gene fusions, gene expression, growth rate in normal and castrated mice, therapeutic response, and experimental metastasis. RESULTS This PCPDX has a mutation in TP53 and loss of PTEN and RB1. We have documented a 100% take rate in mice after thawing the PCPDX tumor from frozen stock. The PCPDX is castrate- and docetaxel-resistant and cisplatin-sensitive, and has gene expression patterns associated with such drug responses. After tail vein injection, the PCPDX tumor cells can colonize the lungs of mice. CONCLUSION This PCPDX, along with others that are established and characterized, will be useful pre-clinically for studying the heterogeneity of prostate cancer biology and testing new therapeutics in models expected to be reflective of the clinical setting.
Collapse
Affiliation(s)
- Brendon M Patierno
- Department of Medicine, Division of Medical Oncology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Wen-Chi Foo
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Tyler Allen
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Jason A Somarelli
- Department of Medicine, Division of Medical Oncology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Kathryn E Ware
- Department of Medicine, Division of Medical Oncology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Santosh Gupta
- Department of Medicine, Division of Medical Oncology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Sandra Wise
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - John P Wise
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Xiaodi Qin
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Dadong Zhang
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Lingfan Xu
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Yanjing Li
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Xufeng Chen
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Brant A Inman
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Shannon J McCall
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Jiaoti Huang
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Rick A Kittles
- Division of Health Equities, Department of Population Sciences, City of Hope, Duarte, 91010, CA, USA
| | - Kouros Owzar
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Simon Gregory
- Department of Medicine, Division of Medical Oncology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Center for Genomics and Computational Biology, Duke University, Durham, NC, 27710, USA
| | - Andrew J Armstrong
- Department of Medicine, Division of Medical Oncology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Daniel J George
- Department of Medicine, Division of Medical Oncology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Steven R Patierno
- Department of Medicine, Division of Medical Oncology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - David S Hsu
- Department of Medicine, Division of Medical Oncology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Center for Genomics and Computational Biology, Duke University, Durham, NC, 27710, USA
| | - Jennifer A Freedman
- Department of Medicine, Division of Medical Oncology, Duke University School of Medicine, Durham, NC, 27710, USA.
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA.
| |
Collapse
|
9
|
Arenas-Gallo C, Owiredu J, Weinstein I, Lewicki P, Basourakos SP, Vince R, Al Hussein Al Awamlh B, Schumacher FR, Spratt DE, Barbieri CE, Shoag JE. Race and prostate cancer: genomic landscape. Nat Rev Urol 2022; 19:547-561. [PMID: 35945369 DOI: 10.1038/s41585-022-00622-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2022] [Indexed: 11/09/2022]
Abstract
In the past 20 years, new insights into the genomic pathogenesis of prostate cancer have been provided. Large-scale integrative genomics approaches enabled researchers to characterize the genetic and epigenetic landscape of prostate cancer and to define different molecular subclasses based on the combination of genetic alterations, gene expression patterns and methylation profiles. Several molecular drivers of prostate cancer have been identified, some of which are different in men of different races. However, the extent to which genomics can explain racial disparities in prostate cancer outcomes is unclear. Future collaborative genomic studies overcoming the underrepresentation of non-white patients and other minority populations are essential.
Collapse
Affiliation(s)
- Camilo Arenas-Gallo
- Department of Urology, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Jude Owiredu
- Department of Urology, NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, USA
| | - Ilon Weinstein
- Department of Urology, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Patrick Lewicki
- Department of Urology, NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, USA
| | - Spyridon P Basourakos
- Department of Urology, NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, USA
| | - Randy Vince
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Bashir Al Hussein Al Awamlh
- Department of Urology, NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, USA.,Department of Urology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Fredrick R Schumacher
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Daniel E Spratt
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Christopher E Barbieri
- Department of Urology, NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, USA
| | - Jonathan E Shoag
- Department of Urology, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA. .,Department of Urology, NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, USA. .,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
10
|
Developing New Treatment Options for Castration-Resistant Prostate Cancer and Recurrent Disease. Biomedicines 2022; 10:biomedicines10081872. [PMID: 36009418 PMCID: PMC9405166 DOI: 10.3390/biomedicines10081872] [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: 06/14/2022] [Revised: 07/20/2022] [Accepted: 07/29/2022] [Indexed: 11/16/2022] Open
Abstract
Prostate cancer (PCa) is a major diagnosed cancer among men globally, and about 20% of patients develop metastatic prostate cancer (mPCa) in the initial diagnosis. PCa is a typical androgen-dependent disease; thus, hormonal therapy is commonly used as a standard care for mPCa by inhibiting androgen receptor (AR) activities, or androgen metabolism. Inevitably, almost all PCa will acquire resistance and become castration-resistant PCa (CRPC) that is associated with AR gene mutations or amplification, the presence of AR variants, loss of AR expression toward neuroendocrine phenotype, or other hormonal receptors. Treating CRPC poses a great challenge to clinicians. Research efforts in the last decade have come up with several new anti-androgen agents to prolong overall survival of CRPC patients. In addition, many potential targeting agents have been at the stage of being able to translate many preclinical discoveries into clinical practices. At this juncture, it is important to highlight the emerging strategies including small-molecule inhibitors to AR variants, DNA repair enzymes, cell survival pathway, neuroendocrine differentiation pathway, radiotherapy, CRPC-specific theranostics and immune therapy that are underway or have recently been completed.
Collapse
|
11
|
Shen D, Peng H, Xia C, Deng Z, Tong X, Wang G, Qian K. The Role of Long Non-Coding RNAs in Epithelial-Mesenchymal Transition-Related Signaling Pathways in Prostate Cancer. Front Mol Biosci 2022; 9:939070. [PMID: 35923466 PMCID: PMC9339612 DOI: 10.3389/fmolb.2022.939070] [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: 05/08/2022] [Accepted: 06/10/2022] [Indexed: 11/17/2022] Open
Abstract
Prostate cancer (PCa) is one of the most common male malignancies with frequent remote invasion and metastasis, leading to high mortality. Epithelial-mesenchymal transition (EMT) is a fundamental process in embryonic development and plays a key role in tumor proliferation, invasion and metastasis. Numerous long non-coding RNAs (lncRNAs) could regulate the occurrence and development of EMT through various complex molecular mechanisms involving multiple signaling pathways in PCa. Given the importance of EMT and lncRNAs in the progression of tumor metastasis, we recapitulate the research progress of EMT-related signaling pathways regulated by lncRNAs in PCa, including AR signaling, STAT3 signaling, Wnt/β-catenin signaling, PTEN/PI3K/AKT signaling, TGF-β/Smad and NF-κB signaling pathways. Furthermore, we summarize four modes of how lncRNAs participate in the EMT process of PCa via regulating relevant signaling pathways.
Collapse
Affiliation(s)
- Dexin Shen
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China
| | - Hongwei Peng
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Caixia Xia
- President’s Office, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhao Deng
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xi Tong
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Gang Wang
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
- Human Genetic Resource Preservation Center of Hubei Province, Wuhan, China
- *Correspondence: Gang Wang, ; Kaiyu Qian,
| | - Kaiyu Qian
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
- Human Genetic Resource Preservation Center of Hubei Province, Wuhan, China
- *Correspondence: Gang Wang, ; Kaiyu Qian,
| |
Collapse
|
12
|
Qian C, Li D, Chen Y. ETS factors in prostate cancer. Cancer Lett 2022; 530:181-189. [PMID: 35033589 PMCID: PMC8832285 DOI: 10.1016/j.canlet.2022.01.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 01/01/2022] [Accepted: 01/10/2022] [Indexed: 12/21/2022]
Abstract
The ETS family of proteins consists of 28 transcription factors, many of which play critical roles in both normal tissue development and homeostasis and have been implicated in development and progression of a variety of cancers. In prostate cancer, gene fusion and overexpression of ETS factors ERG, FLI1, ETV1, ETV4 and ETV5 have been found in half of prostate cancer patients in Caucasian men and define the largest genetic subtype of prostate cancer. This review summarizes the data on the discovery, modeling, molecular taxonomy, lineage plasticity and therapeutic targeting of ETS family members in prostate cancer.
Collapse
Affiliation(s)
- Cheng Qian
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA; Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Dan Li
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Yu Chen
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, NY, 10065, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA.
| |
Collapse
|
13
|
Hidden clues in prostate cancer - Lessons learned from clinical and pre-clinical approaches on diagnosis and risk stratification. Cancer Lett 2022; 524:182-192. [PMID: 34687792 DOI: 10.1016/j.canlet.2021.10.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 09/17/2021] [Accepted: 10/13/2021] [Indexed: 12/18/2022]
Abstract
The heterogeneity of prostate cancer is evident at clinical, morphological and molecular levels. To aid clinical decision making, a three-tiered system for risk stratification is used to designate low-, intermediate-, and high-risk of disease progression. Intermediate-risk prostate cancers are the most frequently diagnosed, and even with common diagnostic features, can exhibit vastly different clinical progression. Thus, improved risk stratification methods are needed to better predict patient outcomes. Here, we provide an overview of the improvements in diagnosis/prognosis arising from advances in pathology reporting of prostate cancer, which can improve risk stratification, especially for patients with intermediate-risk disease. This review discusses updates to pathology reporting of morphological growth patterns, and proposes the utility of integrating prognostic biomarkers or innovative imaging techniques to enhance clinical decision-making. To complement clinical studies, experimental approaches using patient-derived tumors have highlighted important cellular and morphological features associated with aggressive disease that may impact treatment response. The intersection of urology, pathology and scientific disciplines is required to work towards a common goal of understanding disease pathogenesis, improving the stratification of patients with intermediate-risk disease and subsequently defining optimal treatment strategies using precision-based approaches.
Collapse
|
14
|
Bernasocchi T, Theurillat JPP. SPOP-mutant prostate cancer: Translating fundamental biology into patient care. Cancer Lett 2021; 529:11-18. [PMID: 34974131 DOI: 10.1016/j.canlet.2021.12.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 11/30/2021] [Accepted: 12/21/2021] [Indexed: 02/06/2023]
Abstract
Comprehensive cancer genome studies have revealed genetically-defined subtypes of prostate cancer with distinct truncal driver mutations. Because prostate cancer has been largely seen as a rather uniform disease, the clinical significance of this discovery remained largely obscure. However, recent findings imply distinct biological features and therapeutic vulnerabilities linked to specific truncal mutations. Here we review our current understanding of prostate cancers harboring recurrent point mutations in the ubiquitin ligase adaptor protein SPOP and discuss opportunities for future clinical translation. More specifically, activation of the androgen receptor (AR) signaling emerges as the key oncogenic pathway. SPOP-mutant prostate cancer patients respond to AR inhibition in various clinical settings. Molecular insights on how mutant SPOP promotes tumorigenesis may open more specific therapeutic avenues which, in combination with conventional AR-targeting agents, could improve the outcome of patients with SPOP-mutant prostate cancer.
Collapse
Affiliation(s)
- Tiziano Bernasocchi
- Institute of Oncology Research, Bellinzona, TI, 6500, Switzerland; Università della Svizzera italiana (USI), Faculty of Biomedical Sciences, TI, 6900, Lugano, Switzerland
| | - Jean-Philippe P Theurillat
- Institute of Oncology Research, Bellinzona, TI, 6500, Switzerland; Università della Svizzera italiana (USI), Faculty of Biomedical Sciences, TI, 6900, Lugano, Switzerland.
| |
Collapse
|
15
|
Shao L, Wang J, Karatas O, Ittmann M. MEX3D is an oncogenic driver in prostate cancer. Prostate 2021; 81:1202-1213. [PMID: 34455614 PMCID: PMC8460603 DOI: 10.1002/pros.24216] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 01/10/2021] [Accepted: 02/19/2021] [Indexed: 11/05/2022]
Abstract
BACKGROUND Prostate cancer (PCa) is the most common visceral malignancy and the second leading cause of cancer deaths in US men. The two most common genetic alterations in PCa are expression of the TMPRSS2/ERG (TE) fusion gene and loss of the PTEN tumor suppressor. These genetic alterations act cooperatively to transform prostatic epithelium but the exact mechanisms involved are unclear. METHODS Microarray expression analysis of immortalized prostate epithelial cells transformed by loss of PTEN and expression of the TE fusion revealed MEX3D as one of the most highly upregulated genes. MEX3D expression in prostate cancer was examined in patient samples and in silico. In vitro and in vivo studies to characterize the biological impact of MEX3D were carried out. Analysis of the TCGA PanCancer database revealed TCF3 as a major target of MEX3D. The induction of TCF3 by MEX3D was confirmed and the biological impact of TCF3 examined by in vitro studies. RESULTS MEX3D is expressed at increased levels in prostate cancer and is increased by decreased PTEN and/or expression of the TE fusion gene and drives soft agar colony formation, invasion and tumor formation in vivo. The known oncogenic transcription factor TCF3 is highly correlated with MEX3D in prostate cancer. MEX3D expression strongly induces TCF3, which promotes soft agar colony formation and invasion in vitro. CONCLUSIONS Loss of PTEN and expression of the TE fusion gene in prostate cancer strongly upregulates expression of MEX3D and its target TCF3 and promotes transformation associated phenotypes via this pathway.
Collapse
Affiliation(s)
- Longjiang Shao
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA
- Michael E. DeBakey Dept. of Veterans Affairs Medical Center, Houston, Texas, USA
| | - Jianghua Wang
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA
- Michael E. DeBakey Dept. of Veterans Affairs Medical Center, Houston, Texas, USA
| | - Omer Karatas
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA
- Michael E. DeBakey Dept. of Veterans Affairs Medical Center, Houston, Texas, USA
| | - Michael Ittmann
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA
- Michael E. DeBakey Dept. of Veterans Affairs Medical Center, Houston, Texas, USA
| |
Collapse
|
16
|
Imada EL, Sanchez DF, Dinalankara W, Vidotto T, Ebot EM, Tyekucheva S, Franco GR, Mucci LA, Loda M, Schaeffer EM, Lotan T, Marchionni L. Transcriptional landscape of PTEN loss in primary prostate cancer. BMC Cancer 2021; 21:856. [PMID: 34311724 PMCID: PMC8314517 DOI: 10.1186/s12885-021-08593-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/06/2021] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND PTEN is the most frequently lost tumor suppressor in primary prostate cancer (PCa) and its loss is associated with aggressive disease. However, the transcriptional changes associated with PTEN loss in PCa have not been described in detail. In this study, we highlight the transcriptional changes associated with PTEN loss in PCa. METHODS Using a meta-analysis approach, we leveraged two large PCa cohorts with experimentally validated PTEN and ERG status by Immunohistochemistry (IHC), to derive a transcriptomic signature of PTEN loss, while also accounting for potential confounders due to ERG rearrangements. This signature was expanded to lncRNAs using the TCGA quantifications from the FC-R2 expression atlas. RESULTS The signatures indicate a strong activation of both innate and adaptive immune systems upon PTEN loss, as well as an expected activation of cell-cycle genes. Moreover, we made use of our recently developed FC-R2 expression atlas to expand this signature to include many non-coding RNAs recently annotated by the FANTOM consortium. Highlighting potential novel lncRNAs associated with PTEN loss and PCa progression. CONCLUSION We created a PCa specific signature of the transcriptional landscape of PTEN loss that comprises both the coding and an extensive non-coding counterpart, highlighting potential new players in PCa progression. We also show that contrary to what is observed in other cancers, PTEN loss in PCa leads to increased activation of the immune system. These findings can help the development of new biomarkers and help guide therapy choices.
Collapse
Affiliation(s)
- Eddie Luidy Imada
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| | | | - Wikum Dinalankara
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thiago Vidotto
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ericka M Ebot
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Svitlana Tyekucheva
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Gloria Regina Franco
- Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Lorelei Ann Mucci
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | - Tamara Lotan
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Luigi Marchionni
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
17
|
McKay RR, Xie W, Ye H, Fennessy FM, Zhang Z, Lis R, Calagua C, Rathkopf D, Laudone VP, Bubley GJ, Einstein DJ, Chang PK, Wagner AA, Parsons JK, Preston MA, Kilbridge K, Chang SL, Choudhury AD, Pomerantz MM, Trinh QD, Kibel AS, Taplin ME. Results of a Randomized Phase II Trial of Intense Androgen Deprivation Therapy prior to Radical Prostatectomy in Men with High-Risk Localized Prostate Cancer. J Urol 2021; 206:80-87. [PMID: 33683939 PMCID: PMC9807004 DOI: 10.1097/ju.0000000000001702] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE This multicenter randomized phase 2 trial investigates the impact of intense androgen deprivation on radical prostatectomy pathologic response and radiographic and tissue biomarkers in localized prostate cancer (NCT02903368). MATERIALS AND METHODS Eligible patients had a Gleason score ≥4+3=7, prostate specific antigen >20 ng/mL or T3 disease and lymph nodes <20 mm. In Part 1, patients were randomized 1:1 to apalutamide, abiraterone acetate, prednisone and leuprolide (AAPL) or abiraterone, prednisone, leuprolide (APL) for 6 cycles (1 cycle=28 days) followed by radical prostatectomy. Surgical specimens underwent central review. The primary end point was the rate of pathologic complete response or minimum residual disease (minimum residual disease, tumor ≤5 mm). Secondary end points included prostate specific antigen response, positive margin rate and safety. Magnetic resonance imaging and tissue biomarkers of pathologic outcomes were explored. RESULTS The study enrolled 118 patients at 4 sites. Median age was 61 years and 94% of patients had high-risk disease. The combined pathologic complete response or minimum residual disease rate was 22% in the AAPL arm and 20% in the APL arm (difference: 1.5%; 1-sided 95% CI -11%, 14%; 1-sided p=0.4). No new safety signals were observed. There was low concordance and correlation between posttherapy magnetic resonance imaging assessed and pathologically assessed tumor volume. PTEN-loss, ERG positivity and presence of intraductal carcinoma were associated with extensive residual tumor. CONCLUSIONS Intense neoadjuvant hormone therapy in high-risk prostate cancer resulted in favorable pathologic responses (tumor <5 mm) in 21% of patients. Pathologic responses were similar between treatment arms. Part 2 of this study will investigate the impact of adjuvant hormone therapy on biochemical recurrence.
Collapse
Affiliation(s)
- Rana R. McKay
- University of California San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093-0987
| | - Wanling Xie
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215
| | - Huihui Ye
- University of California Los Angeles, Los Angeles, CA 90095
| | - Fiona M. Fennessy
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215
| | - Zhenwei Zhang
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215
| | - Rosina Lis
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215
| | - Carla Calagua
- University of California Los Angeles, Los Angeles, CA 90095
| | - Dana Rathkopf
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065
| | - Vincent P. Laudone
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065
| | - Glenn J. Bubley
- Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA 02215
| | - David J. Einstein
- Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA 02215
| | - Peter K. Chang
- Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA 02215
| | - Andrew A. Wagner
- Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA 02215
| | - J. Kellogg Parsons
- University of California San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093-0987
| | - Mark A. Preston
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215
| | - Kerry Kilbridge
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215
| | - Steven L. Chang
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215
| | | | - Mark M. Pomerantz
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215
| | - Quoc-Dien Trinh
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215
| | - Adam S. Kibel
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215
| | - Mary-Ellen Taplin
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215
| |
Collapse
|
18
|
Lu Z, Song W, Zhang Y, Wu C, Zhu M, Wang H, Li N, Zhou Y, Xu H. Combined Anti-Cancer Effects of Platycodin D and Sorafenib on Androgen-Independent and PTEN-Deficient Prostate Cancer. Front Oncol 2021; 11:648985. [PMID: 34026624 PMCID: PMC8138035 DOI: 10.3389/fonc.2021.648985] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 04/08/2021] [Indexed: 12/15/2022] Open
Abstract
Castration-resistant (androgen-independent) and PTEN-deficient prostate cancer is a challenge in clinical practice. Sorafenib has been recommended for the treatment of this type of cancer, but is associated with several adverse effects. Platycodin D (PD) is a triterpene saponin with demonstrated anti-cancer effects and a good safety profile. Previous studies have indicated that PC3 cells (PTEN -/-, AR -/-) are sensitive to PD, suggesting that it may also be a useful treatment for castration-resistance prostate cancer. We herein investigated the effects of combining PD with sorafenib to treat PTEN-deficient prostate cancer cells. Our data show that PD promotes sorafenib-induced apoptosis and cell cycle arrest in PC3 cells. Of interest, PD only promoted the anti-cancer effects of sorafenib in Akt-positive and PTEN-negative prostate cancer cells. Mechanistic studies revealed that PD promoted p-Akt ubiquitination by increasing the p-Akt level. PD also increased the protein and mRNA expression of FOXO3a, the downstream target of Akt. Meanwhile, PD promoted the activity of FOXO3a and increased the protein expression of Fasl, Bim and TRAIL. Interestingly, when FOXO3a expression was inhibited, the antitumor effects of both PD and sorafenib were individually inhibited, and the more potent effects of the combination treatment were inhibited. Thus, the combination of PD and sorafenib may exert potent anti-cancer effects specifically via FOXO3a. The use of Akt inhibitors or FOXO3a agonists, such as PD, may represent a promising approach for the treatment of androgen-independent and PTEN-deficient prostate cancer.
Collapse
Affiliation(s)
- Zongliang Lu
- Department of Clinical Nutrition, Daping Hospital, Army Medical University, Chongqing, China
| | - Wei Song
- Department of Clinical Nutrition, Daping Hospital, Army Medical University, Chongqing, China
| | - Yaowen Zhang
- Department of Clinical Nutrition, Daping Hospital, Army Medical University, Chongqing, China
| | - Changpeng Wu
- Department of Clinical Nutrition, Daping Hospital, Army Medical University, Chongqing, China
| | - Mingxing Zhu
- Department of Clinical Nutrition, Daping Hospital, Army Medical University, Chongqing, China
| | - He Wang
- Department of Clinical Nutrition, Daping Hospital, Army Medical University, Chongqing, China
| | - Na Li
- Department of Clinical Nutrition, Daping Hospital, Army Medical University, Chongqing, China
| | - Yong Zhou
- Department of Clinical Nutrition, Banan District People's Hospital of Chongqing, Chongqing, China
| | - Hongxia Xu
- Department of Clinical Nutrition, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
19
|
Eerola SK, Kohvakka A, Tammela TLJ, Koskinen PJ, Latonen L, Visakorpi T. Expression and ERG regulation of PIM kinases in prostate cancer. Cancer Med 2021; 10:3427-3436. [PMID: 33932111 PMCID: PMC8124112 DOI: 10.1002/cam4.3893] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/04/2021] [Accepted: 03/23/2021] [Indexed: 11/26/2022] Open
Abstract
The three oncogenic PIM family kinases have been implicated in the development of prostate cancer (PCa). The aim of this study was to examine the mRNA and protein expression levels of PIM1, PIM2, and PIM3 in PCa and their associations with the MYC and ERG oncogenes. We utilized prostate tissue specimens of normal, benign prostatic hyperplasia (BPH), prostatic intraepithelial neoplasia (PIN), untreated PCa, and castration‐resistant prostate cancer (CRPC) for immunohistochemical (IHC) analysis. In addition, we analyzed data from publicly available mRNA expression and chromatin immunoprecipitation sequencing (ChIP‐Seq) datasets. Our data demonstrated that PIM expression levels are significantly elevated in PCa compared to benign samples. Strikingly, the expression of both PIM1 and PIM2 was further increased in CRPC compared to PCa. We also demonstrated a significant association between upregulated PIM family members and both the ERG and MYC oncoproteins. Interestingly, ERG directly binds to the regulatory regions of all PIM genes and upregulates their expression. Furthermore, ERG suppression with siRNA reduced the expression of PIM in PCa cells. These results provide evidence for cooperation of PIM and the MYC and ERG oncoproteins in PCa development and progression and may help to stratify suitable patients for PIM‐targeted therapies.
Collapse
Affiliation(s)
- Sini K Eerola
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, Tampere University Hospital, Tampere, Finland
| | - Annika Kohvakka
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, Tampere University Hospital, Tampere, Finland
| | - Teuvo L J Tammela
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, Tampere University Hospital, Tampere, Finland.,Department of Urology, Tampere University Hospital, Tampere, Finland
| | | | - Leena Latonen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Tapio Visakorpi
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, Tampere University Hospital, Tampere, Finland.,Fimlab Laboratories Ltd, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
20
|
Segura-Moreno YY, Sanabria-Salas MC, Varela R, Mesa JA, Serrano ML. Decoding the heterogeneous landscape in the development prostate cancer. Oncol Lett 2021; 21:376. [PMID: 33777200 PMCID: PMC7988715 DOI: 10.3892/ol.2021.12637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 06/02/2020] [Indexed: 01/02/2023] Open
Abstract
Prostate cancer (PCa) is characterized as being histologically and molecularly heterogeneous; however, this is not only incorrect among individuals, but also at the multiple foci level, which originates in the prostate gland itself. The reasons for such heterogeneity have not been fully elucidated; however, understanding these may be crucial in determining the course of the disease. PCa is characterized by a complex network of chromosomal rearrangements, which simultaneously deregulate multiple genes; this could explain the appearance of exclusive events associated with molecular subtypes, which have been extensively investigated to establish clinical management and the development of therapies targeted to this type of cancer. From a clinical aspect, the prognosis of the patient has focused on the characteristics of the index lesion (the largest focus in PCa); however, a significant percentage of patients (11%) also exhibit an aggressive secondary foci, which may determine the prognosis of the disease, and could be the determining factor of why, in different studies, the classification of the subtypes does not have an association with prognosis. Due to the aforementioned reasons, the analysis of molecular subtypes in several foci, from the same individual could assist in determining the association between clinical evolution and management of patients with PCa. Castration-resistant PCa (CRPC) has the worst prognosis and develops following androgen ablation therapy. Currently, there are two models to explain the development of CRPC: i) The selection model and ii) the adaptation model; both of which, have been found to include alterations described in the molecular subtypes, such as Enhancer of zeste 2 polycomb repressive complex 2 subunit overexpression, isocitrate dehydrogenase (NAPD+)1 and forkhead box A1 mutations, suggesting that the presence of specific molecular alterations could predict the development of CRPC. This type of analysis could lead to a biological understanding of PCa, to develop personalized medicine strategies, which could improve the response to treatment thus, avoiding the development of resistance. Therefore, the present review discusses the primary molecular factors, to which variable heterogeneity in PCa progress has been attributed.
Collapse
Affiliation(s)
- Yenifer Yamile Segura-Moreno
- Cancer Biology Research Group, National Institute of Cancerology, Bogota 110411, Colombia.,Department of Chemistry, Faculty of Sciences, National University of Colombia, University City, Bogota 111321, Colombia
| | | | - Rodolfo Varela
- Department of Urology, National Institute of Cancerology, Bogota 110411, Colombia.,Department of Urology, National University of Colombia, University City, Bogota 111321, Colombia
| | - Jorge Andrés Mesa
- Department of Pathology, National Institute of Cancerology, Bogota 110411, Colombia
| | - Martha Lucia Serrano
- Cancer Biology Research Group, National Institute of Cancerology, Bogota 110411, Colombia.,Department of Chemistry, Faculty of Sciences, National University of Colombia, University City, Bogota 111321, Colombia
| |
Collapse
|
21
|
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: 8.0] [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
|
22
|
Chung JS, Morgan TM, Hong SK. Clinical implications of genomic evaluations for prostate cancer risk stratification, screening, and treatment: a narrative review. Prostate Int 2020; 8:99-106. [PMID: 33102389 PMCID: PMC7557186 DOI: 10.1016/j.prnil.2020.09.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 09/06/2020] [Indexed: 02/08/2023] Open
Abstract
New classification systems based on molecular features have been introduced to improve precision medicine for prostate cancer (PCa). This review covers the increasing risk of PCa and the differences in response to targeted therapy that are related to specific gene variations. We believe that genomic evaluations will be useful for guiding PCa risk stratification, screening, and treatment. We searched the PubMed and MEDLINE databases for articles related to genomic testing for PCa that were published in 2020 or earlier. There is increasing evidence that germline mutations in DNA repair genes, such as BRCA1/2 or ATM, are closely related to the development and aggressiveness of PCa. Targeted prostate-specific antigen screening based on the presence of germline alterations in DNA repair genes is recommend to achieve an early diagnosis of PCa. In cases of localized PCa, even if it has a favorable risk classification, patients under active surveillance with these gene alterations are likely to develop aggressive PCa. Thus, active treatment may be preferable to active surveillance for these patients. In cases of metastatic castration–resistant PCa, BRCA1/2 and DNA mismatch repair genes may be useful biomarkers for predicting the response to androgen receptor–targeting agents, poly (ADP-ribose) polymerase inhibitors, platinum chemotherapy, prostate-specific membrane antigen–targeted therapy, immunotherapy, and radium-223. Genomic evaluations may allow for risk stratification of patients with PCa based on their molecular features, which may help guide precision medicine for treating PCa.
Collapse
Affiliation(s)
- Jae-Seung Chung
- Department of Urology, Inje University Haeundae Paik Hospital, Busan, Korea
| | - Todd M Morgan
- Department of Urology, University of Michigan, Rogel Cancer Center, Ann Arbor, MI, USA
| | - Sung Kyu Hong
- Department of Urology, Seoul National University College of Medicine, Seoul, Korea.,Department of Urology, Seoul National University Bundang Hospital, Seongnam-si, Korea
| |
Collapse
|
23
|
Turnham DJ, Bullock N, Dass MS, Staffurth JN, Pearson HB. The PTEN Conundrum: How to Target PTEN-Deficient Prostate Cancer. Cells 2020; 9:E2342. [PMID: 33105713 PMCID: PMC7690430 DOI: 10.3390/cells9112342] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/17/2020] [Accepted: 10/20/2020] [Indexed: 12/17/2022] Open
Abstract
Loss of the tumor suppressor phosphatase and tensin homologue deleted on chromosome 10 (PTEN), which negatively regulates the PI3K-AKT-mTOR pathway, is strongly linked to advanced prostate cancer progression and poor clinical outcome. Accordingly, several therapeutic approaches are currently being explored to combat PTEN-deficient tumors. These include classical inhibition of the PI3K-AKT-mTOR signaling network, as well as new approaches that restore PTEN function, or target PTEN regulation of chromosome stability, DNA damage repair and the tumor microenvironment. While targeting PTEN-deficient prostate cancer remains a clinical challenge, new advances in the field of precision medicine indicate that PTEN loss provides a valuable biomarker to stratify prostate cancer patients for treatments, which may improve overall outcome. Here, we discuss the clinical implications of PTEN loss in the management of prostate cancer and review recent therapeutic advances in targeting PTEN-deficient prostate cancer. Deepening our understanding of how PTEN loss contributes to prostate cancer growth and therapeutic resistance will inform the design of future clinical studies and precision-medicine strategies that will ultimately improve patient care.
Collapse
Affiliation(s)
- Daniel J. Turnham
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (D.J.T.); (N.B.); (M.S.D.)
| | - Nicholas Bullock
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (D.J.T.); (N.B.); (M.S.D.)
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK;
| | - Manisha S. Dass
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (D.J.T.); (N.B.); (M.S.D.)
| | - John N. Staffurth
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK;
| | - Helen B. Pearson
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (D.J.T.); (N.B.); (M.S.D.)
| |
Collapse
|
24
|
Nikhil K, Kamra M, Raza A, Shah K. Negative cross talk between LIMK2 and PTEN promotes castration resistant prostate cancer pathogenesis in cells and in vivo. Cancer Lett 2020; 498:1-18. [PMID: 32931887 DOI: 10.1016/j.canlet.2020.09.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/17/2020] [Accepted: 09/10/2020] [Indexed: 12/18/2022]
Abstract
Androgen deprivation therapy (ADT) and androgen receptor (AR) signaling inhibitors are front-line treatments for highly aggressive prostate cancer. However, prolonged inhibition of AR triggers a compensatory activation of PI3K pathway, most often due to the genomic loss of tumor suppressor PTEN, driving progression to the castration-resistant prostate cancer (CRPC) stage, which has very poor prognosis. We uncovered a novel mechanism of PTEN downregulation triggered by LIMK2, which contributes significantly to CRPC pathogenesis. LIMK2 is a CRPC-specific target. Its depletion fully reverses tumorigenesis in vivo. LIMK2 phosphorylates PTEN at five sites, degrading and inhibiting its activity, thereby driving highly aggressive oncogenic phenotypes in cells and in vivo. PTEN also degrades LIMK2 in a feedback loop, which was confirmed in prostates from PTEN-/- and PTEN+/+ mice. LIMK2 is also the missing link between hypoxia and PTEN degradation in CRPC. This is the first study to show a feedback loop between PTEN and its regulator. Uncovering the LIMK2-PTEN loop provides a powerful therapeutic opportunity to retain the activity and stability of PTEN protein by inhibiting LIMK2, thereby halting the progression to CRPC, ADT-resistance and drug-resistance.
Collapse
Affiliation(s)
- Kumar Nikhil
- Department of Chemistry and Purdue University Center for Cancer Research 560 Oval Drive, West Lafayette, IN, 47907, USA
| | - Mohini Kamra
- Department of Chemistry and Purdue University Center for Cancer Research 560 Oval Drive, West Lafayette, IN, 47907, USA
| | - Asif Raza
- Department of Chemistry and Purdue University Center for Cancer Research 560 Oval Drive, West Lafayette, IN, 47907, USA
| | - Kavita Shah
- Department of Chemistry and Purdue University Center for Cancer Research 560 Oval Drive, West Lafayette, IN, 47907, USA.
| |
Collapse
|
25
|
Clonal evaluation of prostate cancer molecular heterogeneity in biopsy samples by dual immunohistochemistry and dual RNA in situ hybridization. Mod Pathol 2020; 33:1791-1801. [PMID: 32238875 DOI: 10.1038/s41379-020-0525-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 03/09/2020] [Accepted: 03/09/2020] [Indexed: 12/19/2022]
Abstract
Prostate cancer is frequently multifocal. Although there may be morphological variation, the genetic underpinnings of each tumor are not clearly understood. To assess the inter and intra tumor molecular heterogeneity in prostate biopsy samples, we developed a combined immunohistochemistry and RNA in situ hybridization method for the simultaneous evaluation of ERG, SPINK1, ETV1, and ETV4. Screening of 601 biopsy cores from 120 consecutive patients revealed multiple alterations in a mutually exclusive manner in 37% of patients, suggesting multifocal tumors with considerable genetic differences. Furthermore, the incidence of molecular heterogeneity was higher in African Americans patients compared with Caucasian American patients. About 47% of the biopsy cores with discontinuous tumor foci showed clonal differences with distinct molecular aberrations. ERG positivity occurred in low-grade cancer, whereas ETV4 expression was observed mostly in high-grade cancer. Further studies revealed correlation between the incidence of molecular markers and clinical and pathologic findings, suggesting potential implications for diagnostic pathology practice, such as defining dominant tumor nodules and discriminating juxtaposed but molecularly different tumors of different grade patterns.
Collapse
|
26
|
Wang ZC, Li Y, Wang KL, Wang L, You BS, Zhao DF, Liu ZQ, Fang RZ, Wang JQ, Zhang W, Zhang JM, Xu WH. miR-5089-5p suppresses castration-resistant prostate cancer resistance to enzalutamide and metastasis via miR-5089-5p/SPINK1/ MAPK/MMP9 signaling. Aging (Albany NY) 2020; 12:14418-14433. [PMID: 32694237 PMCID: PMC7425449 DOI: 10.18632/aging.103485] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 05/27/2020] [Indexed: 01/19/2023]
Abstract
Whether serine protease inhibitor Kazal type 1 (SPINK1) being associated with enzalutamide (Enz) resistance and metastasis of castration-resistant prostate cancer (CRPC) has not been clear. SPINK1 promoted Enz resistance by upregulating Androgen receptor splicing variant 7 (ARv7), and enhanced the invasion/migration of Enz-resistant cells via ERK/p38/ MMP9 signaling. Furthermore, miR-5089-5p suppressed SPINK1 mRNA through direct binding to its 3'UTR, and reversed its pro-proliferative and pro-metastatic effects. Mice bearing SPINK1-knockdown Enz-resistant PCa tumors showed significantly longer survival compared with those bearing wild-type tumors, while treatment with miR-5089-5p inhibitor abrogated the protective effects of SPINK1 knockdown. Taken together, SPINK1 can be used as a biomarker of resistance to Enz, and the miR-5089-5p/SPINK1/MAPK/MMP9 axis is a suitable therapeutic target against Enz-resistant and metastatic CRPC. Methods: The expression of SPINK1 in Enz-resistant prostate cancer (PCa) cell lines was detected through next-generation sequencing data and metastatic PCa patients. In vivo and in vitro experiments were performed to investigate the role of SPINK1 in Enz-resistance and metastasis.
Collapse
Affiliation(s)
- Zhi-Chao Wang
- Heilongjiang Key Laboratory of Scientific Research in Urology, The Forth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yan Li
- Department of Anesthesia, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Ke-Liang Wang
- Heilongjiang Key Laboratory of Scientific Research in Urology, The Forth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Lu Wang
- Heilongjiang Key Laboratory of Scientific Research in Urology, The Forth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Bo-Sen You
- Heilongjiang Key Laboratory of Scientific Research in Urology, The Forth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Dan-Feng Zhao
- Heilongjiang Key Laboratory of Scientific Research in Urology, The Forth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Zhong-Qing Liu
- Heilongjiang Key Laboratory of Scientific Research in Urology, The Forth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Rui-Zhe Fang
- Heilongjiang Key Laboratory of Scientific Research in Urology, The Forth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Jia-Qi Wang
- Heilongjiang Key Laboratory of Scientific Research in Urology, The Forth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Wei Zhang
- Heilongjiang Key Laboratory of Scientific Research in Urology, The Forth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Jin-Ming Zhang
- Heilongjiang Key Laboratory of Scientific Research in Urology, The Forth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Wan-Hai Xu
- Heilongjiang Key Laboratory of Scientific Research in Urology, The Forth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| |
Collapse
|
27
|
Leflunomide triggers synthetic lethality in PTEN-deficient prostate cancer. Prostate Cancer Prostatic Dis 2020; 23:718-723. [PMID: 32661432 PMCID: PMC7666085 DOI: 10.1038/s41391-020-0251-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/08/2020] [Accepted: 06/30/2020] [Indexed: 11/08/2022]
Abstract
BACKGROUND The loss of PTEN function presents in up to 50% of late-stage prostate cancers, and is therefore a potential target for therapeutics. PTEN-deficient cells depend on de novo pyrimidine synthesis, a feature that can present a vulnerability. METHODS We utilized in vitro growth assays and in vivo xenograft models to test the effect of de novo pyrimidine synthesis inhibition on prostate cell lines. RESULTS Here, we demonstrate that PTEN-deficient prostate cancer cell lines are susceptible to inhibition of de novo pyrimidine synthesis by leflunomide. Tumor growth inhibition was observed in vitro and in vivo following leflunomide treatment, and is likely due to an overwhelming accumulation of DNA damage. CONCLUSIONS Our work highlights that synthetic lethality arises upon the combination of PTEN loss and leflunomide treatment in prostate cancer, and may present a therapeutic opportunity for this patient population.
Collapse
|
28
|
Wang M, Chen Y, Cai W, Feng H, Du T, Liu W, Jiang H, Pasquarelli A, Weizmann Y, Wang X. In situ self-assembling Au-DNA complexes for targeted cancer bioimaging and inhibition. Proc Natl Acad Sci U S A 2020; 117:308-316. [PMID: 31843938 PMCID: PMC6955332 DOI: 10.1073/pnas.1915512116] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Cancer remains one of the most challenging diseases to treat. For accurate cancer diagnosis and targeted therapy, it is important to assess the localization of the affected area of cancers. The general approaches for cancer diagnostics include pathological assessments and imaging. However, these methods only generally assess the tumor area. In this study, by taking advantage of the unique microenvironment of cancers, we effectively utilize in situ self-assembled biosynthetic fluorescent gold nanocluster-DNA (GNC-DNA) complexes to facilitate safe and targeted cancer theranostics. In in vitro and in vivo tumor models, our self-assembling biosynthetic approach allowed for precise bioimaging and inhibited cancer growth after one injection of DNA and gold precursors. These results demonstrate that in situ bioresponsive self-assembling GNC-PTEN (phosphatase and tensin homolog) complexes could be an effective noninvasive technique for accurate cancer bioimaging and treatment, thus providing a safe and promising cancer theranostics platform for cancer therapy.
Collapse
Affiliation(s)
- Maonan Wang
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Laboratory), School of Biological Science and Medical Engineering, Southeast University, 210096 Nanjing, China
| | - Yun Chen
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Laboratory), School of Biological Science and Medical Engineering, Southeast University, 210096 Nanjing, China
| | - Weijuan Cai
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Laboratory), School of Biological Science and Medical Engineering, Southeast University, 210096 Nanjing, China
| | - Huan Feng
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Laboratory), School of Biological Science and Medical Engineering, Southeast University, 210096 Nanjing, China
| | - Tianyu Du
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Laboratory), School of Biological Science and Medical Engineering, Southeast University, 210096 Nanjing, China
| | - Weiwei Liu
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Laboratory), School of Biological Science and Medical Engineering, Southeast University, 210096 Nanjing, China
| | - Hui Jiang
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Laboratory), School of Biological Science and Medical Engineering, Southeast University, 210096 Nanjing, China
| | - Alberto Pasquarelli
- Institute of Electron Devices and Circuits, Ulm University, 89069 Ulm, Germany
| | - Yossi Weizmann
- Department of Chemistry, Ben-Gurion University of the Negev, 8410501 Beer-Sheva, Israel
| | - Xuemei Wang
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Laboratory), School of Biological Science and Medical Engineering, Southeast University, 210096 Nanjing, China;
| |
Collapse
|
29
|
Pulido R, Mingo J, Gaafar A, Nunes-Xavier CE, Luna S, Torices L, Angulo JC, López JI. Precise Immunodetection of PTEN Protein in Human Neoplasia. Cold Spring Harb Perspect Med 2019; 9:cshperspect.a036293. [PMID: 31501265 DOI: 10.1101/cshperspect.a036293] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PTEN is a major tumor-suppressor protein whose expression and biological activity are frequently diminished in sporadic or inherited cancers. PTEN gene deletion or loss-of-function mutations favor tumor cell growth and are commonly found in clinical practice. In addition, diminished PTEN protein expression is also frequently observed in tumor samples from cancer patients in the absence of PTEN gene alterations. This makes PTEN protein levels a potential biomarker parameter in clinical oncology, which can guide therapeutic decisions. The specific detection of PTEN protein can be achieved by using highly defined anti-PTEN monoclonal antibodies (mAbs), characterized with precision in terms of sensitivity for the detection technique, specificity for PTEN binding, and constraints of epitope recognition. This is especially relevant taking into consideration that PTEN is highly targeted by mutations and posttranslational modifications, and different PTEN protein isoforms exist. The precise characterization of anti-PTEN mAb reactivity is an important step in the validation of these reagents as diagnostic and prognostic tools in clinical oncology, including their routine use in analytical immunohistochemistry (IHC). Here, we review the current status on the use of well-defined anti-PTEN mAbs for PTEN immunodetection in the clinical context and discuss their potential usefulness and limitations for a more precise cancer diagnosis and patient benefit.
Collapse
Affiliation(s)
- Rafael Pulido
- Biocruces Bizkaia Health Research Institute, Barakaldo 48903, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao 48011, Spain
| | - Janire Mingo
- Biocruces Bizkaia Health Research Institute, Barakaldo 48903, Spain
| | - Ayman Gaafar
- Department of Pathology, Cruces University Hospital, Barakaldo 48903, Spain
| | - Caroline E Nunes-Xavier
- Biocruces Bizkaia Health Research Institute, Barakaldo 48903, Spain.,Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo N-0310, Norway
| | - Sandra Luna
- Biocruces Bizkaia Health Research Institute, Barakaldo 48903, Spain
| | - Leire Torices
- Biocruces Bizkaia Health Research Institute, Barakaldo 48903, Spain
| | - Javier C Angulo
- Department of Urology, University Hospital of Getafe, Getafe, Madrid 28904, Spain.,Clinical Department, European University of Madrid, Laureate Universities, Madrid 28904, Spain
| | - José I López
- Biocruces Bizkaia Health Research Institute, Barakaldo 48903, Spain.,Department of Pathology, Cruces University Hospital, Barakaldo 48903, Spain.,University of the Basque Country, Leioa 48940, Spain
| |
Collapse
|
30
|
Hinton JP, Dvorak K, Roberts E, French WJ, Grubbs JC, Cress AE, Tiwari HA, Nagle RB. A Method to Reuse Archived H&E Stained Histology Slides for a Multiplex Protein Biomarker Analysis. Methods Protoc 2019; 2:mps2040086. [PMID: 31731599 PMCID: PMC6960855 DOI: 10.3390/mps2040086] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 11/07/2019] [Accepted: 11/08/2019] [Indexed: 12/13/2022] Open
Abstract
Archived Hematoxylin and Eosin (H&E) stained pathology slides are routinely stored to index formalin-fixed paraffin-embedded (FFPE) sample tissue blocks. FFPE blocks are clinically annotated human tumor specimens that can be valuable in studies decades after the tissue is collected. If stored properly, they have the potential to yield a valuable number of serial sectioned slides for diagnostic or research purposes. However, some retrospective studies are limited in scope because the tissue samples have been depleted or not enough material is available in stored blocks for serial sections. The goal of these studies was to determine if archived H&E-stained slides can be directly reutilized by optimizing methods to de-stain and then re-stain the H&E stained slides to allow the detection of several biomarkers of interest using a conjugated antibody with chromogen multiplex immunohistochemistry procedure. This simple but innovative procedure, combined with image analysis techniques, demonstrates the ability to perform precise detection of relevant markers correlated to disease progression in initially identified tumor regions in tissue. This may add clinical value in retaining H&E slides for further use.
Collapse
Affiliation(s)
- James P. Hinton
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona Cancer Center, Tucson, AZ 85724, USA;
- Ventana/Roche Tissue Diagnostics, Tucson, AZ 85755, USA; (K.D.); (E.R.); (W.J.F.); (J.C.G.)
| | - Katerina Dvorak
- Ventana/Roche Tissue Diagnostics, Tucson, AZ 85755, USA; (K.D.); (E.R.); (W.J.F.); (J.C.G.)
| | - Esteban Roberts
- Ventana/Roche Tissue Diagnostics, Tucson, AZ 85755, USA; (K.D.); (E.R.); (W.J.F.); (J.C.G.)
| | - Wendy J. French
- Ventana/Roche Tissue Diagnostics, Tucson, AZ 85755, USA; (K.D.); (E.R.); (W.J.F.); (J.C.G.)
| | - Jon C. Grubbs
- Ventana/Roche Tissue Diagnostics, Tucson, AZ 85755, USA; (K.D.); (E.R.); (W.J.F.); (J.C.G.)
| | - Anne E. Cress
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona Cancer Center, Tucson, AZ 85724, USA;
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, USA
- Correspondence: ; Tel.: +1-520-626-7553
| | - Hina A. Tiwari
- Department of Medical Imaging, College of Medicine, University of Arizona, Tucson, AZ 85724, USA;
| | - Raymond B. Nagle
- Department of Pathology, College of Medicine, the University of Arizona, Tucson, AZ 85724, USA;
| |
Collapse
|
31
|
Chen WS, Aggarwal R, Zhang L, Zhao SG, Thomas GV, Beer TM, Quigley DA, Foye A, Playdle D, Huang J, Lloyd P, Lu E, Sun D, Guan X, Rettig M, Gleave M, Evans CP, Youngren J, True L, Lara P, Kothari V, Xia Z, Chi KN, Reiter RE, Maher CA, Feng FY, Small EJ, Alumkal JJ. Genomic Drivers of Poor Prognosis and Enzalutamide Resistance in Metastatic Castration-resistant Prostate Cancer. Eur Urol 2019; 76:562-571. [PMID: 30928160 PMCID: PMC6764911 DOI: 10.1016/j.eururo.2019.03.020] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 03/13/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND Metastatic castration-resistant prostate cancer (mCRPC) is the lethal form of the disease. Several recent studies have identified genomic alterations in mCRPC, but the clinical implications of these genomic alterations have not been fully elucidated. OBJECTIVE To use whole-genome sequencing (WGS) to assess the association between key driver gene alterations and overall survival (OS), and to use whole-transcriptome RNA sequencing to identify genomic drivers of enzalutamide resistance. DESIGN, SETTING, AND PARTICIPANTS We performed survival analyses and gene set enrichment analysis (GSEA) on WGS and RNA sequencing results for a cohort of 101 mCRPC patients. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS OS was the clinical endpoint for all univariate and multivariable survival analyses. Candidate drivers of enzalutamide resistance were identified in an unbiased manner, and mutations of the top candidate were further assessed for enrichment among enzalutamide-resistant patients using Fisher's exact test. RESULTS AND LIMITATIONS Harboring two DNA alterations in RB1 was independently predictive of poor OS (median 14.1 vs 42.0mo; p=0.007) for men with mCRPC. GSEA identified the Wnt/β-catenin pathway as the top differentially modulated pathway among enzalutamide-resistant patients. Furthermore, β-catenin mutations were exclusive to enzalutamide-resistant patients (p=0.01) and independently predictive of poor OS (median 13.6 vs 41.7mo; p=0.025). CONCLUSIONS The presence of two RB1 DNA alterations identified in our WGS analysis was independently associated with poor OS among men with mCRPC. The Wnt/β-catenin pathway plays an important role in enzalutamide resistance, with differential pathway expression and enrichment of β-catenin mutations in enzalutamide-resistant patients. Moreover, β-catenin mutations were predictive of poor OS in our cohort. PATIENT SUMMARY We observed a correlation between genomic findings for biopsy samples from metastases from men with metastatic castration-resistant prostate cancer (mCRPC) and clinical outcomes. This work sheds new light on clinically relevant genomic alterations in mCRPC and provides a roadmap for the development of new personalized treatment regimens in mCRPC.
Collapse
Affiliation(s)
- William S Chen
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA; Yale School of Medicine, New Haven, CT, USA
| | - Rahul Aggarwal
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA; Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Li Zhang
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA; Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | | | - George V Thomas
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Tomasz M Beer
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - David A Quigley
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Adam Foye
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA; Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Denise Playdle
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA; Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | | | - Paul Lloyd
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA; Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Eric Lu
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Duanchen Sun
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Xiangnan Guan
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Matthew Rettig
- University of California Los Angeles, Los Angeles, CA, USA; VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | | | | | - Jack Youngren
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA; Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | | | - Primo Lara
- University of California Davis, Davis, CA, USA
| | - Vishal Kothari
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Zheng Xia
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Kim N Chi
- University of British Columbia, Vancouver, Canada
| | | | | | - Felix Y Feng
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA; Departments of Radiation Oncology and Urology, University of California San Francisco, San Francisco, CA, USA
| | - Eric J Small
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA; Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
| | - Joshi J Alumkal
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | | |
Collapse
|
32
|
Hamid AA, Gray KP, Huang Y, Bowden M, Pomerantz M, Loda M, Sweeney CJ. Loss of PTEN Expression Detected by Fluorescence Immunohistochemistry Predicts Lethal Prostate Cancer in Men Treated with Prostatectomy. Eur Urol Oncol 2019; 2:475-482. [DOI: 10.1016/j.euo.2018.09.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 09/09/2018] [Indexed: 01/10/2023]
|
33
|
Pseudogene Associated Recurrent Gene Fusion in Prostate Cancer. Neoplasia 2019; 21:989-1002. [PMID: 31446281 PMCID: PMC6713813 DOI: 10.1016/j.neo.2019.07.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/22/2019] [Accepted: 07/23/2019] [Indexed: 02/08/2023] Open
Abstract
We present the functional characterization of a pseudogene associated recurrent gene fusion in prostate cancer. The fusion gene KLK4-KLKP1 is formed by the fusion of the protein coding gene KLK4 with the noncoding pseudogene KLKP1. Screening of a cohort of 659 patients (380 Caucasian American; 250 African American, and 29 patients from other races) revealed that the KLK4-KLKP1 is expressed in about 32% of prostate cancer patients. Correlative analysis with other ETS gene fusions and SPINK1 revealed a concomitant expression pattern of KLK4-KLKP1 with ERG and a mutually exclusive expression pattern with SPINK1, ETV1, ETV4, and ETV5. Development of an antibody specific to KLK4-KLKP1 fusion protein confirmed the expression of the full-length KLK4-KLKP1 protein in prostate tissues. The in vitro and in vivo functional assays to study the oncogenic properties of KLK4-KLKP1 confirmed its role in cell proliferation, cell invasion, intravasation, and tumor formation. Presence of strong ERG and AR binding sites located at the fusion junction in KLK4-KLKP1 suggests that the fusion gene is regulated by ERG and AR. Correlative analysis of clinical data showed an association of KLK4-KLKP1 with lower preoperative PSA values and in young men (<50 years) with prostate cancer. Screening of patient urine samples showed that KLK4-KLKP1 can be detected noninvasively in urine. Taken together, we present KLK4-KLKP1 as a class of pseudogene associated fusion transcript in cancer with potential applications as a biomarker for routine screening of prostate cancer.
Collapse
|
34
|
Rezk M, Chandra A, Addis D, Møller H, Youssef M, Dasgupta P, Yamamoto H. ETS-related gene ( ERG) expression as a predictor of oncological outcomes in patients with high-grade prostate cancer treated with primary androgen deprivation therapy: a cohort study. BMJ Open 2019; 9:e025161. [PMID: 30852544 PMCID: PMC6429920 DOI: 10.1136/bmjopen-2018-025161] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVES To determine whether ETS-related gene (ERG) expression can be used as a biomarker to predict biochemical recurrence and prostate cancer-specific death in patients with high Gleason grade prostate cancer treated with androgen deprivation therapy (ADT) as monotherapy. METHODS A multicentre retrospective cohort study identifying 149 patients treated with primary ADT for metastatic or non-metastatic prostate cancer with Gleason score 8-10 between 1999 and 2006. Patients planned for adjuvant radiotherapy at diagnosis were excluded. Age at diagnosis, ethnicity, prostate-specific antigen and Charlson-comorbidity score were recorded. Prostatic tissue acquired at biopsy or transurethral resection surgery was assessed for immunohistochemical expression of ERG. Failure of ADT defined as prostate specific antigen nadir +2. Vital status and death certification data determined using the UK National Cancer Registry. Primary outcome measures were overall survival (OS) and prostate cancer specific survival (CSS). Secondary outcome was biochemical recurrence-free survival (BRFS). RESULTS The median OS of our cohort was 60.2 months (CI 52.0 to 68.3). ERG expression observed in 51/149 cases (34%). Multivariate Cox proportional hazards analysis showed no significant association between ERG expression and OS (p=0.41), CSS (p=0.92) and BRFS (p=0.31). Cox regression analysis showed Gleason score (p=0.003) and metastatic status (p<1×10-5) to be the only significant predictors of prostate CSS. CONCLUSIONS No significant association was found between ERG status and any of our outcome measures. Despite a limited sample size, our results suggest that ERG does not appear to be a useful biomarker in predicting response to ADT in patients with high risk prostate cancer.
Collapse
Affiliation(s)
- Mark Rezk
- NIHR Biomedical Research Centre, Kings College London, London, UK
- Intensive Care Unit, Torbay and South Devon NHS Foundation Trust, Torquay, UK
| | - Ashish Chandra
- Department of Histopathology and Cytology, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
| | - Daniel Addis
- Department of Histopathology and Cytology, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
| | - Henrik Møller
- Department of Cancer Epidemiology & Populational Health, Kings College London, London, UK
| | - Mina Youssef
- General Surgery Department, Norfolk and Norwich University Hospitals NHS Foundation Trust, Norwich, UK
- Surgical Oncology Department, National Cancer Institute, Cairo, Egypt
| | - Prokar Dasgupta
- NIHR Biomedical Research Centre, Kings College London, London, UK
| | - Hide Yamamoto
- Department of Urology, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
- Department of Urology, Maidstone Hospital, Maidstone, UK
| |
Collapse
|
35
|
Nicholas TR, Strittmatter BG, Hollenhorst PC. Oncogenic ETS Factors in Prostate Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1210:409-436. [PMID: 31900919 DOI: 10.1007/978-3-030-32656-2_18] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Prostate cancer is unique among carcinomas in that a fusion gene created by a chromosomal rearrangement is a common driver of the disease. The TMPRSS2/ERG rearrangement drives aberrant expression of the ETS family transcription factor ERG in 50% of prostate tumors. Similar rearrangements promote aberrant expression of the ETS family transcription factors ETV1 and ETV4 in another 10% of cases. Together, these three ETS factors are thought to promote tumorigenesis in the majority of prostate cancers. A goal of precision medicine is to be able to apply targeted therapeutics that are specific to disease subtypes. ETS gene rearrangement positive tumors represent the largest molecular subtype of prostate cancer, but to date there is no treatment specific to this marker. In this chapter we will review the latest findings regarding the molecular mechanisms of ETS factor function in the prostate. These molecular details may provide a path towards new therapeutic targets for this subtype of prostate cancer. Further, we will describe efforts to target the oncogenic functions of ETS family transcription factors directly as well as indirectly.
Collapse
Affiliation(s)
| | - Brady G Strittmatter
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN, USA
| | - Peter C Hollenhorst
- Medical Sciences, Indiana University School of Medicine, Bloomington, IN, USA.
| |
Collapse
|
36
|
Comparing Prognostic Utility of a Single-marker Immunohistochemistry Approach with Commercial Gene Expression Profiling Following Radical Prostatectomy. Eur Urol 2018; 74:668-675. [DOI: 10.1016/j.eururo.2018.08.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/13/2018] [Indexed: 11/18/2022]
|
37
|
Shinde D, Albino D, Zoma M, Mutti A, Mapelli SN, Civenni G, Kokanovic A, Merulla J, Perez-Escuredo J, Costales P, Morìs F, Catapano CV, Carbone GM. Transcriptional Reprogramming and Inhibition of Tumor-propagating Stem-like Cells by EC-8042 in ERG-positive Prostate Cancer. Eur Urol Oncol 2018; 2:415-424. [PMID: 31277777 DOI: 10.1016/j.euo.2018.08.024] [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] [Received: 08/16/2018] [Accepted: 08/24/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND The TMPRSS2-ERG gene fusion is the most frequent genetic rearrangement in prostate cancers and results in broad transcriptional reprogramming and major phenotypic changes. Interaction and cooperation of ERG and SP1 may be instrumental in sustaining the tumorigenic and metastatic phenotype and could represent a potential vulnerability in ERG fusion-positive tumors. OBJECTIVE To test the activity of EC-8042, a compound able to block SP1, in cellular and mouse models of ERG-positive prostate cancer. DESIGN, SETTING, AND PARTICIPANTS We evaluated the activity of EC-8042 in cell cultures and ERG/PTEN transgenic/knockout mice that provide reliable models for testing novel therapeutics in this specific disease context. Using a new protocol to generate tumor spheroids from ERG/PTEN mice, we also examined the effects of EC-8042 on tumor-propagating stem-like cancer cells with high self-renewal and tumorigenic capabilities. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The efficacy of EC-8042 was determined by measuring the proliferative capacity and target gene expression in cell cultures, invasive and metastatic capabilities in chick chorioallantoic membrane assays, and tumor development in mice. Significance was determined using statistical test. RESULTS AND LIMITATIONS EC-8042 blocked transcription of ERG-regulated genes and reverted the invasive and metastatic phenotype of VCaP cells. EC-8042 blocked the expansion of stem-like tumor cells in tumor spheroids from VCaP cells and mouse-derived tumors. In ERG/PTEN mice, systemic treatment with EC-8042 inhibited ERG-regulated gene transcription, tumor progression, and tumor-propagating stem-like tumor cells. CONCLUSIONS Our data support clinical testing of EC-8042 for the treatment of ERG-positive prostate cancer in precision medicine approaches. PATIENT SUMMARY In this study, EC-8042, a novel compound with a favorable pharmacological and toxicological profile, exhibited relevant activity in cell cultures and in vivo in a genetically engineered mouse model that closely recapitulates the features of clinically aggressive ERG-positive prostate cancer. Our data indicate that further evaluation of EC-8042 in clinical trials is warranted.
Collapse
Affiliation(s)
- Dheeraj Shinde
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Domenico Albino
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Marita Zoma
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Azzurra Mutti
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Sarah N Mapelli
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Gianluca Civenni
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Aleksandra Kokanovic
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Jessica Merulla
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | | | | | | | - Carlo V Catapano
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Giuseppina M Carbone
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland.
| |
Collapse
|
38
|
Carneiro A, Barbosa ÁRG, Takemura LS, Kayano PP, Moran NKS, Chen CK, Wroclawski ML, Lemos GC, da Cunha IW, Obara MT, Tobias-Machado M, Sowalsky AG, Bianco B. The Role of Immunohistochemical Analysis as a Tool for the Diagnosis, Prognostic Evaluation and Treatment of Prostate Cancer: A Systematic Review of the Literature. Front Oncol 2018; 8:377. [PMID: 30280090 PMCID: PMC6153326 DOI: 10.3389/fonc.2018.00377] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 08/23/2018] [Indexed: 12/12/2022] Open
Abstract
Background: Prostate cancer (PCa) is a heterogeneous disease that lends itself toward numerous therapeutic options depending on its risk stratification. One of the greatest challenges in PCa urologic practice is to select patients who should be referred for biopsy and, for those patients who are diagnosed with cancer, to differentiate between patients with indolent disease from those with an unfavorable prognosis and, to determine ideal patient management and avoid unnecessary interventions. Accordingly, there is a growing body of literature reporting immunohistochemical studies with the objective of determining a prostate cancer prognosis. Among the most frequent biomarkers studied are Ki-67, p53, PTEN, MYC, and ERG. Based on these findings, we systematically reviewed articles that assessed the role of these main prognostic markers in prostate cancer. Methods: Consistent with PRISMA guidelines, we performed a systematic literature search throughout the Web of Science and PubMed Medline databases. We considered all types of studies evaluating the role of Ki-67, p53, PTEN, MYC, and ERG immunohistochemical analysis in prostate cancer until July 2017. Results: We identified 361 articles, 44 of which were summarized in this review. Diagnostically, no single immunohistochemical marker was able to define a tumor as benign or malignant. Prognostically, Ki-67, p53, and MYC were related to the tumor grade given by Gleason score and to the tumor stage (higher levels related to higher tumor grade). Furthermore, Ki-67 was also related to higher PSA levels, shorter disease-free intervals and shorter tumor-specific survival; the latter was also related to p53. The loss of PTEN protein expression showed a higher association with biochemical recurrence and with a worse prognosis, beyond that predicted by the Gleason score and tumor stage. ERG staining also showed a strong association with biochemical recurrence. Conclusion: There are several studies relating immunohistochemical markers with clinical-laboratorial outcomes in prostate cancer, the most frequent being Ki-67, p53, ERG, PTEN, and MYC. However, none of these markers have been validated by literary consensus to be routinely applied in medical practice.
Collapse
Affiliation(s)
- Arie Carneiro
- Department of Urology, Hospital Israelita Albert Einstein, São Paulo, Brazil.,Department of Pathology, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | | | | | | | | | - Carolina Ko Chen
- Department of Urology, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Marcelo Langer Wroclawski
- Department of Urology, Hospital Israelita Albert Einstein, São Paulo, Brazil.,Department of Pathology, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | | | | | - Marcos Takeo Obara
- Department of Pathology, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | | | - Adam G Sowalsky
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Bianca Bianco
- Human Reproduction and Genetics Center, Faculdade de Medicina do ABC, Santo André, Brazil
| |
Collapse
|
39
|
Tabakin AL, Sadimin ET, Tereshchenko I, Kareddula A, Stein MN, Mayer T, Hirshfield KM, Kim IY, Tischfield J, DiPaola RS, Singer EA. Correlation of Prostate Cancer CHD1 Status with Response to Androgen Deprivation Therapy: a Pilot Study. JOURNAL OF GENITOURINARY DISORDERS 2018; 2:1006. [PMID: 30714046 PMCID: PMC6358174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
INTRODUCTION CHD1 has been identified as a tumor suppressor gene in prostate cancer. Previous studies have shown strong associations between CHD1 deletion, prostate specific antigen [PSA] recurrence, and absence of ERG fusion. In this preliminary study we seek to find whether there is an independent correlation between CHD1 status and response to androgen deprivation therapy[ADT]. MATERIALS AND METHODS We identified 11 patients with prostate cancer who underwent prostatectomy and received at least 7 months of ADT at our institution. They were divided into undetectable [PSA < 0.2 ng/mL; n = 8] and detectable [PSA > 0.2 ng/mL; n = 3] according to their serum PSA nadir after 7 months of ADT. Tissue microarray was generated from their formalin-fixed paraffin-embedded prostatectomy and involved lymph node tissues. Fluorescence in situ hybridization [FISH] analysis for CHD1 and immunohistochemical stains for PSA, AR, PTEN, ERG and SPINK1 were performed. RESULTS Our results showed heterogeneity of FISH and immunostains expressions in different foci of tumor. Status of CHD1, ERG, PTEN, or SPINK1 did not correlate with one another or with response to ADT. CONCLUSIONS Additional larger studies may be needed to further elucidate trends between these biomarkers and clinical outcomes in prostate cancer patients.
Collapse
Affiliation(s)
- Alexandra L. Tabakin
- Division of Urologic Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, USA
| | - Evita T. Sadimin
- Section of Pathologic Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, USA
| | - Irina Tereshchenko
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, USA
| | - Aparna Kareddula
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, USA
| | - Mark N. Stein
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, USA
| | - Tina Mayer
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, USA
| | - Kim M. Hirshfield
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, USA
| | - Isaac Y. Kim
- Division of Urologic Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, USA
| | - Jay Tischfield
- Department of Genetics, Human Genetics Institute of New Jersey and Rutgers University, USA
| | - Robert S. DiPaola
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, USA
| | - Eric A. Singer
- Division of Urologic Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, USA
| |
Collapse
|
40
|
Kallio HML, Hieta R, Latonen L, Brofeldt A, Annala M, Kivinummi K, Tammela TL, Nykter M, Isaacs WB, Lilja HG, Bova GS, Visakorpi T. Constitutively active androgen receptor splice variants AR-V3, AR-V7 and AR-V9 are co-expressed in castration-resistant prostate cancer metastases. Br J Cancer 2018; 119:347-356. [PMID: 29988112 PMCID: PMC6070921 DOI: 10.1038/s41416-018-0172-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/08/2018] [Accepted: 06/13/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND A significant subset of prostate cancer (PC) patients with a castration-resistant form of the disease (CRPC) show primary resistance to androgen receptor (AR)-targeting drugs developed against CRPC. As one explanation could be the expression of constitutively active androgen receptor splice variants (AR-Vs), our current objectives were to study AR-Vs and other AR aberrations to better understand the emergence of CRPC. METHODS We analysed specimens from different stages of prostate cancer by next-generation sequencing and immunohistochemistry. RESULTS AR mutations and copy number variations were detected only in CRPC specimens. Genomic structural rearrangements of AR were observed in 5/30 metastatic CRPC patients, but they were not associated with expression of previously known AR-Vs. The predominant AR-Vs detected were AR-V3, AR-V7 and AR-V9, with the expression levels being significantly higher in CRPC cases compared to prostatectomy samples. Out of 25 CRPC metastases that expressed any AR variant, 17 cases harboured expression of all three of these AR-Vs. AR-V7 protein expression was highly heterogeneous and higher in CRPC compared to hormone-naïve tumours. CONCLUSIONS AR-V3, AR-V7 and AR-V9 are co-expressed in CRPC metastases highlighting the fact that inhibiting AR function via regions common to all AR-Vs is likely to provide additional benefit to patients with CRPC.
Collapse
Affiliation(s)
- Heini M L Kallio
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland.
| | - Reija Hieta
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Leena Latonen
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Anniina Brofeldt
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Matti Annala
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Kati Kivinummi
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Teuvo L Tammela
- Department of Urology, University of Tampere, Tampere University Hospital, Tampere, Finland
| | - Matti Nykter
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - William B Isaacs
- The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hans G Lilja
- Departments of Surgery (Urology), Laboratory Medicine and Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.,Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK.,Department of Translational Medicine, Lund University, Malmö, Sweden.,Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - G Steven Bova
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Tapio Visakorpi
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland.,Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
41
|
Rescigno P, Lorente D, Dolling D, Ferraldeschi R, Rodrigues DN, Riisnaes R, Miranda S, Bianchini D, Zafeiriou Z, Sideris S, Ferreira A, Figueiredo I, Sumanasuriya S, Mateo J, Perez-Lopez R, Sharp A, Tunariu N, de Bono JS. Docetaxel Treatment in PTEN- and ERG-aberrant Metastatic Prostate Cancers. Eur Urol Oncol 2018; 1:71-77. [PMID: 29911685 PMCID: PMC5995869 DOI: 10.1016/j.euo.2018.02.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND Loss of PTEN is a common genomic aberration in castration-resistant prostate cancer (CRPC) and is frequently concurrent with ERG rearrangements, causing resistance to next-generation hormonal treatment (NGHT) including abiraterone. The relationship between PTEN loss and docetaxel sensitivity remains uncertain. OBJECTIVE To study the antitumor activity of docetaxel in metastatic CRPC in relation to PTEN and ERG aberrations. DESIGN SETTING AND PARTICIPANTS Single-centre, retrospective analysis of PTEN loss and ERG expression using a previously described immunohistochemistry (IHC) binary classification system. Patients received docetaxel between January 1, 2006 and July 31, 2016. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Response correlations were analyzed using Pearson's χ2 tests and independent-sample t tests. Overall (OS) and progression-free survival (PFS) were analyzed using univariate and multivariate (MVA) Cox regression and Kaplan-Meier methods. RESULTS AND LIMITATIONS Overall, 215 patients were eligible. Established metastatic CRPC prognostic factors were well balanced between PTEN loss (39%) and normal patients (61%). PTEN loss was associated with shorter median OS (25.4 vs 34.7 mo; hazard ratio [HR] 1.66, 95% confidence interval [CI] 1.18-2.13; p = 0.001). There were no differences in median PFS (8.0 vs 9.1 mo; univariate HR 1.20, 95% CI 0.86-1.68; p = 0.28) and PSA response (53.4% vs 50.6%; p = 0.74). PTEN loss was an independent prognostics factor in MVA. ERG status was available for 100 patients. ERG positivity was not associated with OS or PFS. Limitations include the retrospective nature and the single-centre analysis. CONCLUSIONS Our findings suggest that metastatic CRPC with PTEN loss might benefit more from docetaxel than from NGHT. PATIENT SUMMARY In this study we found that metastatic prostate cancer with loss of the PTEN switch may benefit more from docetaxel than from abiraterone.
Collapse
Affiliation(s)
- Pasquale Rescigno
- The Institute of Cancer Research, Sutton, UK; Department of Clinical Medicine and Surgery, Department of Translational Medical Sciences, AOU Federico II, Naples, Italy
| | - David Lorente
- Medical Oncology Service, Hospital Universitario La Fe, Valencia, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Adam Sharp
- The Institute of Cancer Research, Sutton, UK
| | | | | |
Collapse
|
42
|
Sainio M, Visakorpi T, Tolonen T, Ilvesaro J, Bova GS. Expression of neuroendocrine differentiation markers in lethal metastatic castration-resistant prostate cancer. Pathol Res Pract 2018; 214:848-856. [PMID: 29728311 DOI: 10.1016/j.prp.2018.04.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 04/09/2018] [Accepted: 04/20/2018] [Indexed: 12/31/2022]
Abstract
Neuroendocrine differentiation (NED) is a common phenomenon in prostate cancer, and it has been associated with poor prognosis in some studies of primary prostate cancer. Incidence and patterns of NED in metastatic prostate cancer sites have not been examined widely. In this study, we studied expression of three commonly used markers of NED (chromogranin A, neuron specific enolase and synaptophysin) in 89 metastases from 31 men that died of castration-resistant prostate cancer and underwent rapid autopsy, and in 89 hormone-naïve primary tumors removed by radical prostatectomy. In addition, we examined NED association with androgen receptor, ERG and Ki-67 expression in metastatic tumor sites. Morphologically, 1 of 31 cases was classified as small cell carcinoma, and the remaining 30 were classified as usual prostate adenocarcinoma using a recently proposed classification of prostate cancers with NED. Metastases showed more expression of neuron specific enolase and synaptophysin compared to prostatectomies (6.3% of cells vs. 1.0%, p < 0.001 and 4.0% vs. 0.4%, p < 0.001, respectively). At least focal expression of one of the markers was seen in 78% of metastases. Strong expression was relatively uncommon, seen in 3/89 (chromogranin A), 8/89 (neuron specific enolase), and 5/89 (synaptophysin) metastases. Expression of chromogranin A and synaptophysin correlated with each other (r = 0.64, p < 0.001), but expression of neuron specific enolase did not correlate with the two other markers. Extent of NED varied significantly between different metastatic sites in individual patients. Absent androgen receptor expression was associated with strong expression of chromogranin A (p = .02) and neuron specific enolase (p = .02), but not with focal expression of any marker. No clear association was found between expression of NE markers and ERG or Ki-67. In conclusion, NED is a common and heterogeneous phenomenon in metastatic, castration-resistant prostate cancer. NED is more often present in castration-resistant prostate cancer compared to hormone-naïve disease, and it is associated with androgen receptor negativity. More research is needed to understand significance of NED in the progression of prostate cancer.
Collapse
Affiliation(s)
- Miika Sainio
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, FI-33014, Finland.
| | - Tapio Visakorpi
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, FI-33014, Finland.
| | - Teemu Tolonen
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, FI-33014, Finland; Department of Pathology, Tampere University Hospital, Fimlab Laboratories, Tampere, Finland.
| | - Joanna Ilvesaro
- Department of Pathology, Tampere University Hospital, Fimlab Laboratories, Tampere, Finland.
| | - G Steven Bova
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, FI-33014, Finland.
| |
Collapse
|
43
|
Shimazaki J, Chung LWK, Zhau HE, Ichikawa T. Dr. Coffey's visionary contributions to urological research in China and Japan. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2018; 6:15-22. [PMID: 29666826 PMCID: PMC5902716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 03/19/2018] [Indexed: 06/08/2023]
Affiliation(s)
- Jun Shimazaki
- Department of Urology, Graduate School of Medicine, Chiba UniversityChiba, Japan
| | - Leland WK Chung
- Department of Medicine and Surgery, Cedars-Sinai Medical CenterLos Angeles, CA. 90048, USA
| | - Haiyen E Zhau
- Department of Medicine and Surgery, Cedars-Sinai Medical CenterLos Angeles, CA. 90048, USA
| | - Tomohiko Ichikawa
- Department of Urology, Graduate School of Medicine, Chiba UniversityChiba, Japan
| |
Collapse
|
44
|
Smits M, Mehra N, Sedelaar M, Gerritsen W, Schalken JA. Molecular biomarkers to guide precision medicine in localized prostate cancer. Expert Rev Mol Diagn 2018. [PMID: 28635333 DOI: 10.1080/14737159.2017.1345627] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Major advances through tumor profiling technologies, that include next-generation sequencing, epigenetic, proteomic and transcriptomic methods, have been made in primary prostate cancer, providing novel biomarkers that may guide precision medicine in the near future. Areas covered: The authors provided an overview of novel molecular biomarkers in tissue, blood and urine that may be used as clinical tools to assess prognosis, improve selection criteria for active surveillance programs, and detect disease relapse early in localized prostate cancer. Expert commentary: Active surveillance (AS) in localized prostate cancer is an accepted strategy in patients with very low-risk prostate cancer. Many more patients may benefit from watchful waiting, and include patients of higher clinical stage and grade, however selection criteria have to be optimized and early recognition of transformation from localized to lethal disease has to be improved by addition of molecular biomarkers. The role of non-invasive biomarkers is challenging the need for repeat biopsies, commonly performed at 1 and 4 years in men under AS programs.
Collapse
Affiliation(s)
- Minke Smits
- a Department of Urology and Oncology , Radboud Universiteit , Nijmegen , The Netherlands
| | - Niven Mehra
- a Department of Urology and Oncology , Radboud Universiteit , Nijmegen , The Netherlands
| | - Michiel Sedelaar
- a Department of Urology and Oncology , Radboud Universiteit , Nijmegen , The Netherlands
| | - Winald Gerritsen
- a Department of Urology and Oncology , Radboud Universiteit , Nijmegen , The Netherlands
| | - Jack A Schalken
- a Department of Urology and Oncology , Radboud Universiteit , Nijmegen , The Netherlands
| |
Collapse
|
45
|
Latonen L, Afyounian E, Jylhä A, Nättinen J, Aapola U, Annala M, Kivinummi KK, Tammela TTL, Beuerman RW, Uusitalo H, Nykter M, Visakorpi T. Integrative proteomics in prostate cancer uncovers robustness against genomic and transcriptomic aberrations during disease progression. Nat Commun 2018; 9:1176. [PMID: 29563510 PMCID: PMC5862881 DOI: 10.1038/s41467-018-03573-6] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 02/21/2018] [Indexed: 01/23/2023] Open
Abstract
To understand functional consequences of genetic and transcriptional aberrations in prostate cancer, the proteomic changes during disease formation and progression need to be revealed. Here we report high-throughput mass spectrometry on clinical tissue samples of benign prostatic hyperplasia (BPH), untreated primary prostate cancer (PC) and castration resistant prostate cancer (CRPC). Each sample group shows a distinct protein profile. By integrative analysis we show that, especially in CRPC, gene copy number, DNA methylation, and RNA expression levels do not reliably predict proteomic changes. Instead, we uncover previously unrecognized molecular and pathway events, for example, several miRNA target correlations present at protein but not at mRNA level. Notably, we identify two metabolic shifts in the citric acid cycle (TCA cycle) during prostate cancer development and progression. Our proteogenomic analysis uncovers robustness against genomic and transcriptomic aberrations during prostate cancer progression, and significantly extends understanding of prostate cancer disease mechanisms. Understanding of molecular events in cancer requires proteome-level characterisation. Here, proteome profiling of patient samples representing primary and progressed prostate cancer enables the authors to identify pathway alterations that are not reflected at the genomic and transcriptomic levels.
Collapse
Affiliation(s)
- Leena Latonen
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, 33014, Finland.,FimLab Laboratories, Tampere University Hospital, Tampere, 33101, Finland
| | - Ebrahim Afyounian
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, 33014, Finland
| | - Antti Jylhä
- Department of Ophthalmology, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, 33014, Finland
| | - Janika Nättinen
- Department of Ophthalmology, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, 33014, Finland
| | - Ulla Aapola
- Department of Ophthalmology, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, 33014, Finland
| | - Matti Annala
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, 33014, Finland
| | - Kati K Kivinummi
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, 33014, Finland
| | - Teuvo T L Tammela
- Department of Urology, University of Tampere and Tampere University Hospital, Tampere, 33521, Finland
| | - Roger W Beuerman
- Department of Ophthalmology, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, 33014, Finland.,Singapore Eye Research Institute, Singapore, 169856, Singapore.,Duke-NUS Neuroscience, Singapore, 169857, Singapore.,Duke-NUS Medical School Ophthalmology and Visual Sciences Academic Clinical Program, Singapore, 169857, Singapore.,Ophthalmology, Yong Loo Lin Medical School, National University of Singapore, Singapore, 119228, Singapore
| | - Hannu Uusitalo
- Department of Ophthalmology, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, 33014, Finland.,Tays Eye Centre, Tampere University Hospital, Tampere, 33521, Finland
| | - Matti Nykter
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, 33014, Finland. .,Science Center, Tampere University Hospital, Tampere, 33521, Finland.
| | - Tapio Visakorpi
- Prostate Cancer Research Center, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, 33014, Finland. .,FimLab Laboratories, Tampere University Hospital, Tampere, 33101, Finland.
| |
Collapse
|
46
|
Profiling Prostate Cancer Therapeutic Resistance. Int J Mol Sci 2018; 19:ijms19030904. [PMID: 29562686 PMCID: PMC5877765 DOI: 10.3390/ijms19030904] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 03/16/2018] [Accepted: 03/16/2018] [Indexed: 02/06/2023] Open
Abstract
The major challenge in the treatment of patients with advanced lethal prostate cancer is therapeutic resistance to androgen-deprivation therapy (ADT) and chemotherapy. Overriding this resistance requires understanding of the driving mechanisms of the tumor microenvironment, not just the androgen receptor (AR)-signaling cascade, that facilitate therapeutic resistance in order to identify new drug targets. The tumor microenvironment enables key signaling pathways promoting cancer cell survival and invasion via resistance to anoikis. In particular, the process of epithelial-mesenchymal-transition (EMT), directed by transforming growth factor-β (TGF-β), confers stem cell properties and acquisition of a migratory and invasive phenotype via resistance to anoikis. Our lead agent DZ-50 may have a potentially high efficacy in advanced metastatic castration resistant prostate cancer (mCRPC) by eliciting an anoikis-driven therapeutic response. The plasticity of differentiated prostate tumor gland epithelium allows cells to de-differentiate into mesenchymal cells via EMT and re-differentiate via reversal to mesenchymal epithelial transition (MET) during tumor progression. A characteristic feature of EMT landscape is loss of E-cadherin, causing adherens junction breakdown, which circumvents anoikis, promoting metastasis and chemoresistance. The targetable interactions between androgens/AR and TGF-β signaling are being pursued towards optimized therapeutic regimens for the treatment of mCRPC. In this review, we discuss the recent evidence on targeting the EMT-MET dynamic interconversions to overcome therapeutic resistance in patients with recurrent therapeutically resistant prostate cancer. Exploitation of the phenotypic landscape and metabolic changes that characterize the prostate tumor microenvironment in advanced prostate cancer and consequential impact in conferring treatment resistance are also considered in the context of biomarker discovery.
Collapse
|
47
|
Jamaspishvili T, Berman DM, Ross AE, Scher HI, De Marzo AM, Squire JA, Lotan TL. Clinical implications of PTEN loss in prostate cancer. Nat Rev Urol 2018; 15:222-234. [PMID: 29460925 DOI: 10.1038/nrurol.2018.9] [Citation(s) in RCA: 375] [Impact Index Per Article: 62.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Genomic aberrations of the PTEN tumour suppressor gene are among the most common in prostate cancer. Inactivation of PTEN by deletion or mutation is identified in ∼20% of primary prostate tumour samples at radical prostatectomy and in as many as 50% of castration-resistant tumours. Loss of phosphatase and tensin homologue (PTEN) function leads to activation of the PI3K-AKT (phosphoinositide 3-kinase-RAC-alpha serine/threonine-protein kinase) pathway and is strongly associated with adverse oncological outcomes, making PTEN a potentially useful genomic marker to distinguish indolent from aggressive disease in patients with clinically localized tumours. At the other end of the disease spectrum, therapeutic compounds targeting nodes in the PI3K-AKT-mTOR (mechanistic target of rapamycin) signalling pathway are being tested in clinical trials for patients with metastatic castration-resistant prostate cancer. Knowledge of PTEN status might be helpful to identify patients who are more likely to benefit from these therapies. To enable the use of PTEN status as a prognostic and predictive biomarker, analytically validated assays have been developed for reliable and reproducible detection of PTEN loss in tumour tissue and in blood liquid biopsies. The use of clinical-grade assays in tumour tissue has shown a robust correlation between loss of PTEN and its protein as well as a strong association between PTEN loss and adverse pathological features and oncological outcomes. In advanced disease, assessing PTEN status in liquid biopsies shows promise in predicting response to targeted therapy. Finally, studies have shown that PTEN might have additional functions that are independent of the PI3K-AKT pathway, including those affecting tumour growth through modulation of the immune response and tumour microenvironment.
Collapse
Affiliation(s)
- Tamara Jamaspishvili
- Division of Cancer Biology and Genetics, Cancer Research Institute, Queen's University, Kingston, Ontario, Canada.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - David M Berman
- Division of Cancer Biology and Genetics, Cancer Research Institute, Queen's University, Kingston, Ontario, Canada.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Ashley E Ross
- Department of Urology, Johns Hopkins University, Baltimore, MD, USA
| | - Howard I Scher
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, USA
| | - Angelo M De Marzo
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| | - Jeremy A Squire
- Department of Pathology and Legal Medicine, University of Sao Paulo, Campus Universitario Monte Alegre, Ribeirão Preto, Brazil
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
48
|
Shao L, Wang J, Karatas OF, Feng S, Zhang Y, Creighton CJ, Ittmann M. Fibroblast growth factor receptor signaling plays a key role in transformation induced by the TMPRSS2/ERG fusion gene and decreased PTEN. Oncotarget 2018; 9:14456-14471. [PMID: 29581856 PMCID: PMC5865682 DOI: 10.18632/oncotarget.24470] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 02/03/2018] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer is the most common visceral malignancy and the second leading cause of cancer deaths in US men. Correlative studies in human prostate cancers reveal a frequent association of the TMPRSS2/ERG (TE) fusion gene with loss of PTEN and studies in mouse models reveal that ERG expression and PTEN loss synergistically promote prostate cancer progression. To determine the mechanism by which ERG overexpression and PTEN loss leads to transformation, we overexpressed the TE fusion gene and knocked down PTEN in an immortalized but non-transformed prostate epithelial cell line. We show that ERG overexpression in combination with PTEN loss can transform these immortalized but non-tumorigenic cells, while either alteration alone was not sufficient to fully transform these cells. Expression microarray analysis revealed extensive changes in gene expression in cells expressing the TE fusion with loss of PTEN. Among these gene expression changes was increased expression of multiple FGF ligands and receptors. We show that activation of fibroblast growth factor receptor signaling plays a key role in transformation induced by TE fusion gene expression in association with PTEN loss. In addition, in vitro and in silico analysis reveals PTEN loss is associated with widespread increases in FGF ligands and receptors in prostate cancer. Inhibitors of FGF receptor signaling are currently entering the clinic and our results suggests that FGF receptor signaling is a therapeutic target in cancers with TE fusion gene expression and PTEN loss.
Collapse
Affiliation(s)
- Longjiang Shao
- Deptartment of Pathology & Immunology, Baylor College of Medicine, Houston, Texas 77030, USA.,Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas 77030, USA
| | - Jianghua Wang
- Deptartment of Pathology & Immunology, Baylor College of Medicine, Houston, Texas 77030, USA.,Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas 77030, USA
| | - Omer Faruk Karatas
- Deptartment of Pathology & Immunology, Baylor College of Medicine, Houston, Texas 77030, USA.,Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas 77030, USA
| | - Shu Feng
- Deptartment of Pathology & Immunology, Baylor College of Medicine, Houston, Texas 77030, USA.,Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas 77030, USA
| | - Yiqun Zhang
- Department of Medicine, Baylor College of Medicine, Houston, Texas 77030, USA.,Dan L. Duncan Cancer Center Division of Biostatistics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Chad J Creighton
- Department of Medicine, Baylor College of Medicine, Houston, Texas 77030, USA.,Dan L. Duncan Cancer Center Division of Biostatistics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Michael Ittmann
- Deptartment of Pathology & Immunology, Baylor College of Medicine, Houston, Texas 77030, USA.,Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas 77030, USA
| |
Collapse
|
49
|
Kivinummi K, Urbanucci A, Leinonen K, Tammela TLJ, Annala M, Isaacs WB, Bova GS, Nykter M, Visakorpi T. The expression of AURKA is androgen regulated in castration-resistant prostate cancer. Sci Rep 2017; 7:17978. [PMID: 29269934 PMCID: PMC5740165 DOI: 10.1038/s41598-017-18210-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 12/06/2017] [Indexed: 11/13/2022] Open
Abstract
Although second generation endocrine therapies have significantly improved survival, castration-resistant prostate cancer (CRPC) cells are eventually able to escape available hormonal treatments due to reactivation of androgen receptor (AR) signaling. Identification of novel, non-classical and druggable AR-target genes may provide new approaches to treat CRPC. Our previous analyses suggested that Aurora kinase A (AURKA) is regulated by androgens in prostate cancer cells that express high levels of AR. Here, we provide further evidence that AURKA is significantly overexpressed in AR-positive CRPC samples carrying amplification of AR gene and/or expressing AR in high levels. We also demonstrate androgen-induced AR binding in the intronic region of AURKA. The expression of AURKA is increased upon androgen stimulation in LNCaP-ARhi cells that express high levels of AR. The growth of the cells was also significantly inhibited by an AURKA specific inhibitor, alisertib (MLN8237). Together, these findings suggest that the expression of AURKA is regulated by androgen in prostate cancer cells that highly express AR, emphasizing its potential as a therapeutic target in patients with CRPC.
Collapse
Affiliation(s)
- Kati Kivinummi
- BioMediTech Institute, Faculty of Medicine and Life Sciences, University of Tampere, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland.
| | - Alfonso Urbanucci
- BioMediTech Institute, Faculty of Medicine and Life Sciences, University of Tampere, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland.,Centre for Molecular Medicine Norway, Nordic European Molecular Biology Laboratory Partnership, Forksningsparken, University of Oslo, Oslo, Norway.,Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,K.G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway
| | - Katri Leinonen
- BioMediTech Institute, Faculty of Medicine and Life Sciences, University of Tampere, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| | - Teuvo L J Tammela
- Department of Urology, Tampere University Hospital and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Matti Annala
- BioMediTech Institute, Faculty of Medicine and Life Sciences, University of Tampere, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| | - William B Isaacs
- The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
| | - G Steven Bova
- BioMediTech Institute, Faculty of Medicine and Life Sciences, University of Tampere, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| | - Matti Nykter
- BioMediTech Institute, Faculty of Medicine and Life Sciences, University of Tampere, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| | - Tapio Visakorpi
- BioMediTech Institute, Faculty of Medicine and Life Sciences, University of Tampere, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
50
|
Ullman D, Dorn D, Rais-Bahrami S, Gordetsky J. Clinical Utility and Biologic Implications of Phosphatase and Tensin Homolog (PTEN) and ETS-related Gene (ERG) in Prostate Cancer. Urology 2017; 113:59-70. [PMID: 29225123 DOI: 10.1016/j.urology.2017.11.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 11/15/2017] [Accepted: 11/15/2017] [Indexed: 12/13/2022]
Abstract
Phosphatase and tensin homolog (PTEN) and ETS-related gene (ERG) mutations are commonly found in prostate cancer. Although mouse studies have demonstrated that PTEN and ERG cooperatively interact during tumorigenesis, human studies examining these genes have been inconclusive. A systematic PubMed search including original articles assessing the pathogenesis of PTEN and ERG in prostate cancer was performed. Studies examining ERG's prognostic significance have conflicting results. Studies examining PTEN and ERG simultaneously found these genes are likely to occur together, but cooperative tumorigenesis functions have not been conclusively established. PTEN mutations are associated with a range of prognostic features. However, the practical clinical utility of this information remains to be determined.
Collapse
Affiliation(s)
- David Ullman
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL
| | - David Dorn
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL
| | - Soroush Rais-Bahrami
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL; Department of Radiology, University of Alabama at Birmingham, Birmingham, AL
| | - Jennifer Gordetsky
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL; Department of Urology, University of Alabama at Birmingham, Birmingham, AL.
| |
Collapse
|