1
|
Abikar A, Mustafa MMS, Athalye RR, Nadig N, Tamboli N, Babu V, Keshavamurthy R, Ranganathan P. Comparative transcriptome of normal and cancer-associated fibroblasts. BMC Cancer 2024; 24:1231. [PMID: 39369238 PMCID: PMC11456241 DOI: 10.1186/s12885-024-13006-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/27/2024] [Indexed: 10/07/2024] Open
Abstract
BACKGROUND The characteristics of a tumor are largely determined by its interaction with the surrounding micro-environment (TME). TME consists of both cellular and non-cellular components. Cancer-associated fibroblasts (CAFs) are a major component of the TME. They are a source of many secreted factors that influence the survival and progression of tumors as well as their response to drugs. Identification of markers either overexpressed in CAFs or unique to CAFs would pave the way for novel therapeutic strategies that in combination with conventional chemotherapy are likely to have better patient outcome. METHODS Fibroblasts have been derived from Benign Prostatic Hyperplasia (BPH) and prostate cancer. RNA from these has been used to perform a transcriptome analysis in order to get a comparative profile of normal and cancer-associated fibroblasts. RESULTS The study has identified 818 differentially expressed mRNAs and 17 lincRNAs between normal and cancer-associated fibroblasts. Also, 15 potential lincRNA-miRNA-mRNA combinations have been identified which may be potential biomarkers. CONCLUSIONS This study identified differentially expressed markers between normal and cancer-associated fibroblasts that would help in targeted therapy against CAFs/derived factors, in combination with conventional therapy. However, this would in future need more experimental validation.
Collapse
Affiliation(s)
- Apoorva Abikar
- Centre for Human Genetics, Bengaluru, India
- Manipal Academy of Higher Education, Manipal, India
| | | | | | | | | | - Vinod Babu
- Institute of Nephro-Urology, Bengaluru, India
| | | | - Prathibha Ranganathan
- Centre for Human Genetics, Bengaluru, India.
- Manipal Academy of Higher Education, Manipal, India.
| |
Collapse
|
2
|
Bogaard M, Strømme JM, Kidd SG, Johannessen B, Bakken AC, Lothe RA, Axcrona K, Skotheim RI, Axcrona U. GRIN3A: A biomarker associated with a cribriform pattern and poor prognosis in prostate cancer. Neoplasia 2024; 55:101023. [PMID: 38944914 PMCID: PMC11267071 DOI: 10.1016/j.neo.2024.101023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 06/20/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024]
Abstract
Prostate cancer with a cribriform pattern, including invasive cribriform carcinoma (ICC) and/or intraductal carcinoma (IDC) is associated with a poor prognosis, and the underlying mechanisms are unclear. Therefore, we aimed to identify biomarkers for this feature. Using a radical prostatectomy cohort, we performed within-patient differential expression analyses with RNA sequencing data to compare samples with a cribriform pattern to those with non-cribriform Gleason pattern 4 (NcGP4; n=13). ACSM1, GRIN3A, PCDHB2, and REG4 were identified as differentially expressed, and validation was performed using real-time reverse transcription polymerase chain reaction (n=99; 321 RNA samples) and RNA in situ hybridization on tissue microarrays (n=479; 2047 tissue cores). GRIN3A was significantly higher expressed in cribriform pattern vs. NcGP4, when assessed within the same patient (n=27; p=0.005) and between different patients (n=83; p=0.001). Tissue cores with IDC more often expressed GRIN3A compared to ICC, NcGP4, and benign tissue (52 % vs. ≤ 32 %). When IDC and NcGP4 was compared within the same patient (173 pairs of tissue cores; 54 patients), 38 (22 %) of the tissue microarray core pairs had GRIN3A expression in only IDC, 33 (19 %) had expression in both IDC and NcGP4, 14 (8 %) in only NcGP4 and 88 (51 %) were negative in both entities (p=0.001). GRIN3A was as well associated with biochemical recurrence (log-rank, p=0.002). In conclusion, ectopic GRIN3A expression is an RNA-based biomarker for the presence of cribriform prostate cancer, particularly for IDC.
Collapse
Affiliation(s)
- Mari Bogaard
- Department of Pathology, Oslo University Hospital-Radiumhospitalet, Oslo, Norway; Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Jonas M Strømme
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway; Department of Informatics, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Susanne G Kidd
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Bjarne Johannessen
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| | - Anne C Bakken
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| | - Ragnhild A Lothe
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Karol Axcrona
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway; Department of Urology, Akershus University Hospital, Lørenskog, Norway
| | - Rolf I Skotheim
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway; Department of Informatics, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Ulrika Axcrona
- Department of Pathology, Oslo University Hospital-Radiumhospitalet, Oslo, Norway; Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway.
| |
Collapse
|
3
|
von Eyben FE, Kairemo K, Kapp DS. Prostate-Specific Antigen as an Ultrasensitive Biomarker for Patients with Early Recurrent Prostate Cancer: How Low Shall We Go? A Systematic Review. Biomedicines 2024; 12:822. [PMID: 38672176 PMCID: PMC11048591 DOI: 10.3390/biomedicines12040822] [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: 09/15/2023] [Revised: 12/25/2023] [Accepted: 03/05/2024] [Indexed: 04/28/2024] Open
Abstract
Serum prostate-specific antigen (PSA) needs to be monitored with ultrasensitive PSA assays (uPSAs) for oncologists to be able to start salvage radiotherapy (SRT) while PSA is <0.5 µg/L for patients with prostate cancer (PCa) relapsing after a radical prostatectomy (RP). Our systematic review (SR) aimed to summarize uPSAs for patients with localized PCa. The SR was registered as InPLASY2023110084. We searched for studies on Google Scholar, PUBMED and reference lists of reviews and studies. We only included studies on uPSAs published in English and excluded studies of women, animals, sarcoidosis and reviews. Of the 115 included studies, 39 reported PSA assay methods and 76 reported clinical findings. Of 67,479 patients, 14,965 developed PSA recurrence (PSAR) and 2663 died. Extremely low PSA nadir and early developments of PSA separated PSAR-prone from non-PSAR-prone patients (cumulative p value 3.7 × 1012). RP patients with the lowest post-surgery PSA nadir and patients who had the lowest PSA at SRT had the fewest deaths. In conclusion, PSA for patients with localized PCa in the pre-PSAR phase of PCa is strongly associated with later PSAR and survival. A rising but still exceedingly low PSA at SRT predicts a good 5-year overall survival.
Collapse
Affiliation(s)
| | - Kalevi Kairemo
- Department of Molecular Radiotherapy & Nuclear Medicine, Docrates Cancer Center, FI-00185 Helsinki, Finland;
| | - Daniel S. Kapp
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
4
|
Skotheim RI, Bogaard M, Carm KT, Axcrona U, Axcrona K. Prostate cancer: Molecular aspects, consequences, and opportunities of the multifocal nature. Biochim Biophys Acta Rev Cancer 2024; 1879:189080. [PMID: 38272101 DOI: 10.1016/j.bbcan.2024.189080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 01/27/2024]
Abstract
Prostate cancer is unique compared to other major cancers due to the presence of multiple primary malignant foci in the majority of patients at the time of diagnosis. Each malignant focus has distinct somatic mutations and gene expression patterns, which represents a challenge for the development of prognostic tests for localized prostate cancer. Additionally, the molecular heterogeneity of advanced prostate cancer has important implications for management, particularly for patients with metastatic and locally recurrent cancer. Studies have shown that prostate cancers with mutations in DNA damage response genes are more sensitive to drugs inhibiting the poly ADP-ribose polymerase (PARP) enzyme. However, testing for such mutations should consider both spatial and temporal heterogeneity. Here, we summarize studies where multiregional genomics and transcriptomics analyses have been performed for primary prostate cancer. We further discuss the vast interfocal heterogeneity and how prognostic biomarkers and a molecular definition of the index tumor should be developed. The concept of focal treatments in prostate cancer has been evolving as a demand from patients and clinicians and is one example where there is a need for defining an index tumor. Here, biomarkers must have proven value for individual malignant foci. The potential discovery and implementation of biomarkers that are agnostic to heterogeneity are also explored as an alternative to multisample testing. Thus, deciding upon whole-organ treatment, such as radical prostatectomy, should depend on information from biomarkers which are informative for the whole organ.
Collapse
Affiliation(s)
- Rolf I Skotheim
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Department of Informatics, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway.
| | - Mari Bogaard
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Kristina T Carm
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Ulrika Axcrona
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Karol Axcrona
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Department of Urology, Akershus University Hospital, Lørenskog, Norway
| |
Collapse
|
5
|
Śmieszek A, Marcinkowska K, Małas Z, Sikora M, Kępska M, Nowakowska BA, Deperas M, Smyk M, Rodriguez-Galindo C, Raciborska A. Identification and characterization of stromal-like cells with CD207 +/low CD1a +/low phenotype derived from histiocytic lesions - a perspective in vitro model for drug testing. BMC Cancer 2024; 24:105. [PMID: 38342891 PMCID: PMC10860276 DOI: 10.1186/s12885-023-11807-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 12/28/2023] [Indexed: 02/13/2024] Open
Abstract
BACKGROUND Histiocytoses are rare disorders manifested by increased proliferation of pathogenic myeloid cells sharing histological features with macrophages or dendritic cells and accumulating in various organs, i.a., bone and skin. Pre-clinical in vitro models that could be used to determine molecular pathways of the disease are limited, hence research on histiocytoses is challenging. The current study compares cytophysiological features of progenitor, stromal-like cells derived from histiocytic lesions (sl-pHCs) of three pediatric patients with different histiocytoses types and outcomes. The characterized cells may find potential applications in drug testing. METHODS Molecular phenotype of the cells, i.e. expression of CD1a and CD207 (langerin), was determined using flow cytometry. Cytogenetic analysis included GTG-banded metaphases and microarray (aCGH) evaluation. Furthermore, the morphology and ultrastructure of cells were evaluated using a confocal and scanning electron microscope. The microphotographs from the confocal imaging were used to reconstruct the mitochondrial network and its morphology. Basic cytophysiological parameters, such as viability, mitochondrial activity, and proliferation, were analyzed using multiple cellular assays, including Annexin V/7-AAD staining, mitopotential analysis, BrdU test, clonogenicity analysis, and distribution of cells within the cell cycle. Biomarkers potentially associated with histiocytoses progression were determined using RT-qPCR at mRNA, miRNA and lncRNA levels. Intracellular accumulation of histiocytosis-specific proteins was detected with Western blot. Cytotoxicyty and IC50 of vemurafenib and trametinib were determined with MTS assay. RESULTS Obtained cellular models, i.e. RAB-1, HAN-1, and CHR-1, are heterogenic in terms of molecular phenotype and morphology. The cells express CD1a/CD207 markers characteristic for dendritic cells, but also show intracellular accumulation of markers characteristic for cells of mesenchymal origin, i.e. vimentin (VIM) and osteopontin (OPN). In subsequent cultures, cells remain viable and metabolically active, and the mitochondrial network is well developed, with some distinctive morphotypes noted in each cell line. Cell-specific transcriptome profile was noted, providing information on potential new biomarkers (non-coding RNAs) with diagnostic and prognostic features. The cells showed different sensitivity to vemurafenib and trametinib. CONCLUSION Obtained and characterized cellular models of stromal-like cells derived from histiocytic lesions can be used for studies on histiocytosis biology and drug testing.
Collapse
Affiliation(s)
- Agnieszka Śmieszek
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, Norwida 31, 50-375, Wroclaw, Poland.
| | - Klaudia Marcinkowska
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wroclaw University of Environmental and Life Sciences, Norwida 27B, 50-375, Wroclaw, Poland
| | - Zofia Małas
- Department of Oncology and Surgical Oncology for Children and Youth, Institute of Mother and Child, Kasprzaka 17a, 01-211, Warsaw, Poland
| | - Mateusz Sikora
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wroclaw University of Environmental and Life Sciences, Norwida 27B, 50-375, Wroclaw, Poland
| | - Martyna Kępska
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wroclaw University of Environmental and Life Sciences, Norwida 27B, 50-375, Wroclaw, Poland
| | - Beata A Nowakowska
- Medical Genetics Department, Cytogenetics Laboratory, Institute of Mother and Child, Kasprzaka 17a, 01-211, Warsaw, Poland
| | - Marta Deperas
- Medical Genetics Department, Cytogenetics Laboratory, Institute of Mother and Child, Kasprzaka 17a, 01-211, Warsaw, Poland
| | - Marta Smyk
- Medical Genetics Department, Cytogenetics Laboratory, Institute of Mother and Child, Kasprzaka 17a, 01-211, Warsaw, Poland
| | | | - Anna Raciborska
- Department of Oncology and Surgical Oncology for Children and Youth, Institute of Mother and Child, Kasprzaka 17a, 01-211, Warsaw, Poland.
| |
Collapse
|
6
|
Agbetuyi-Tayo P, Gbadebo M, Rotimi OA, Rotimi SO. Advancements in Biomarkers of Prostate Cancer: A Review. Technol Cancer Res Treat 2024; 23:15330338241290029. [PMID: 39440372 PMCID: PMC11497500 DOI: 10.1177/15330338241290029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/01/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024] Open
Abstract
Prostate cancer (PCa) is one of the most prevalent and deadly cancers among men, particularly affecting men of African descent and contributing significantly to cancer-related morbidity and mortality worldwide. The disease varies widely, from slow-developing forms to highly aggressive or potentially fatal variants. Accurate risk stratification is crucial for making therapeutic decisions and designing adequate clinical trials. This review assesses a broad spectrum of diagnostic and prognostic biomarkers, many of which are incorporated into clinical guidelines, including the Prostate Health Index (PHI), 4Kscore, STHLM3, PCA3, SelectMDx, ExoDx Prostate Intelliscore (EPI), and MiPS. It also highlights emerging biomarkers with preclinical support, such as urinary non-coding RNAs and DNA methylation patterns. Additionally, the review explores the role of tumor-associated microbiota in PCa, offering new insights into its potential contributions to disease understanding. By examining the latest advancements in PCa biomarkers, this review enhances understanding their roles in disease management.
Collapse
Affiliation(s)
- Praise Agbetuyi-Tayo
- Department of Biochemistry, Covenant University, Ota, Nigeria
- Covenant Applied Informatics and Communication Africa Centre of Excellence (CApIC-ACE), Covenant University, Ota, Nigeria
| | - Mary Gbadebo
- Department of Biochemistry, Covenant University, Ota, Nigeria
- Covenant Applied Informatics and Communication Africa Centre of Excellence (CApIC-ACE), Covenant University, Ota, Nigeria
| | - Oluwakemi A. Rotimi
- Department of Biochemistry, Covenant University, Ota, Nigeria
- Covenant Applied Informatics and Communication Africa Centre of Excellence (CApIC-ACE), Covenant University, Ota, Nigeria
| | - Solomon O. Rotimi
- Department of Biochemistry, Covenant University, Ota, Nigeria
- Covenant Applied Informatics and Communication Africa Centre of Excellence (CApIC-ACE), Covenant University, Ota, Nigeria
| |
Collapse
|
7
|
Bogaard M, Skotheim RI, Maltau AV, Kidd SG, Lothe RA, Axcrona K, Axcrona U. 'High proliferative cribriform prostate cancer' defines a patient subgroup with an inferior prognosis. Histopathology 2023; 83:853-869. [PMID: 37501635 DOI: 10.1111/his.15012] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/28/2023] [Accepted: 07/06/2023] [Indexed: 07/29/2023]
Abstract
AIMS A cribriform pattern, reactive stroma (RS), PTEN, Ki67 and ERG are promising prognostic biomarkers in primary prostate cancer (PCa). We aim to determine the relative contribution of these factors and the Cancer of the Prostate Risk Assessment Postsurgical (CAPRA-S) score in predicting PCa prognosis. METHODS AND RESULTS We included 475 patients who underwent radical prostatectomy (2010-12, median follow-up = 8.7 years). Cribriform pattern was identified in 57% of patients, PTEN loss in 55%, ERG expression in 51%, RS in 39% and high Ki67 in 9%. In patients with multiple samples from the same malignant focus and either PTEN loss or high Ki67, intrafocal heterogeneity for PTEN and Ki67 expression was detected in 55% and 89%, respectively. In patients with samples from two or more foci, interfocal heterogeneity was detected in 46% for PTEN and 6% for Ki67. A cribriform pattern and Ki67 were independent predictors of biochemical recurrence (BCR) and clinical recurrence (CR), whereas ERG expression was an independent predictor of CR. Besides CAPRA-S, a cribriform pattern provided the highest relative proportion of explained variation for predicting BCR (11%), and Ki67 provided the highest relative proportion of explained variation for CR (21%). In patients with a cribriform pattern, high Ki67 was associated with a higher risk of BCR [hazard ratio (HR) = 2.83, P < 0.001] and CR (HR = 4.35, P < 0.001). CONCLUSIONS High Ki67 in patients with a cribriform pattern identifies a patient subgroup with particularly poor prognosis, which we termed 'high proliferative cribriform prostate cancer'. These results support reporting a cribriform pattern in pathology reports, and advocate implementing Ki67.
Collapse
Affiliation(s)
- Mari Bogaard
- Department of Pathology, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Rolf I Skotheim
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
- Department of Informatics, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Aase V Maltau
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| | - Susanne G Kidd
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Ragnhild A Lothe
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Karol Axcrona
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
- Department of Urology, Akershus University Hospital, Lørenskog, Norway
| | - Ulrika Axcrona
- Department of Pathology, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| |
Collapse
|
8
|
Li Y, Fang Z, Ge S, Li J, Qu L, Shi X, Zhang W, Sun Y, Ren S, Wang L. Long non-coding RNA ENST00000503625 is a potential prognostic biomarker and metastasis suppressor gene in prostate cancer. J Cancer Res Clin Oncol 2023; 149:7305-7317. [PMID: 36920562 DOI: 10.1007/s00432-023-04676-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 03/04/2023] [Indexed: 03/16/2023]
Abstract
BACKGROUND Dysregulation of Long Non-coding RNAs (lncRNAs) emerges to be a hallmark of cancers. Metastatic prostate cancer and localized disease that recurs after treatment are clinical challenges, it remains unclear how lncRNA plays a role in those processes. METHODS From previous RNA-Seq data on 65 prostate cancer and adjacent normal tissues. We identified a novel lncRNA ENST00000503625 down-regulated in prostate cancer and correlated with tumor progression characteristics. Public datasets were examined for associations between ENST00000503625 expression and clinical parameters and prognoses. Subsequently, we constructed and externally validated a nomogram for predicting biochemical recurrence (BCR). Finally, in vitro experiments were carried out to determine how ENST00000503625 functions biologically in prostate cancer. RESULTS Low ENST00000503625 in tumor was associated with poor clinical features and prognoses. TCGA pan-cancer analysis found that ENST00000503625 was deregulated in a variety of tumors and correlated with overall survival, disease-specific survival, and progression-free survival. The nomogram for predicting BCR was constructed using TCGA data, which exhibited excellent accuracy in external validation with Chinese Prostate Cancer Genome and Epigenome Atlas data. Gene Ontology and KEGG pathway analysis found that genes related to ENST00000503625 were enriched in multiple tumor progression related pathways. When ENST00000503625 was knocked down in vitro, the epithelial-mesenchymal transition was induced, by which cancer cells migrated and invaded more readily. CONCLUSION Our data suggested that ENST00000503625 may serve as a potential prognostic marker or a therapeutic target for prostate cancer metastases.
Collapse
Affiliation(s)
- Yaoming Li
- Department of Urology, Institute of Surgery Research, Daping Hospital/Army Medical Center, Army Medical University, Chongqing, 400042, China
- Department of Urology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Ziyu Fang
- Department of Urology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Silun Ge
- Department of Urology, Jinling Hospital, Nanjing Medical University, Nanjing, 201101, China
| | - Jingyi Li
- Department of Urology, Institute of Surgery Research, Daping Hospital/Army Medical Center, Army Medical University, Chongqing, 400042, China
| | - Le Qu
- Department of Urology, Jinling Hospital, Nanjing Medical University, Nanjing, 201101, China
| | - Xiaolei Shi
- Department of Urology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Wei Zhang
- Department of Urology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yinghao Sun
- Department of Urology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Shancheng Ren
- Department of Urology, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China.
| | - Luofu Wang
- Department of Urology, Institute of Surgery Research, Daping Hospital/Army Medical Center, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
9
|
Carm KT, Johannessen B, Bogaard M, Bakken AC, Maltau AV, Hoff AM, Axcrona U, Axcrona K, Lothe RA, Skotheim RI. Somatic mutations reveal complex metastatic seeding from multifocal primary prostate cancer. Int J Cancer 2023; 152:945-951. [PMID: 35880692 PMCID: PMC10087486 DOI: 10.1002/ijc.34226] [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: 03/14/2022] [Revised: 06/27/2022] [Accepted: 07/05/2022] [Indexed: 01/06/2023]
Abstract
Primary prostate cancer shows a striking intraorgan molecular heterogeneity, with multiple spatially separated malignant foci in the majority of patients. Metastatic prostate cancer, however, typically reveals more homogenous molecular profiles, suggesting a monoclonal origin of the metastatic lesions. Longitudinal mutational spectra, comparing multiple primary lesions with metastases from the same patients remain poorly defined. We have here analyzed somatic mutations in multisampled, spatio-temporal biobanked lesions (38 samples from primary foci and 1 sample from each of 8 metastases from seven prostate cancer patients) applying a custom-designed panel targeting 68 prostate cancer relevant genes. The metastatic samples were taken at time of primary surgery and up to 7 years later, and sampling included circulating tumor DNA in plasma or solid metastatic tissue samples. A total of 282 somatic mutations were detected, with a range of 0 to 25 mutations per sample. Although seven samples had solely private mutations, the remaining 39 samples had both private and shared mutations. Seventy-four percent of mutations in metastases were not found in any primary samples, and vice versa, 96% of mutations in primary cancers were not found in any metastatic samples. However, for three patients, shared mutations were found suggesting the focus of origin, including mutations in AKT1, FOXA1, HOXB13, RB1 and TP53. In conclusion, the spatio-temporal heterogeneous nature of multifocal disease is emphasized in our study, and underlines the importance of testing a recent sample in genomics-based precision medicine for metastatic prostate cancer.
Collapse
Affiliation(s)
- Kristina T Carm
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway.,Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Bjarne Johannessen
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| | - Mari Bogaard
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway.,Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Pathology, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| | - Anne Cathrine Bakken
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| | - Aase V Maltau
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| | - Andreas M Hoff
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| | - Ulrika Axcrona
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway.,Department of Pathology, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| | - Karol Axcrona
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway.,Department of Urology, Akershus University Hospital, Lørenskog, Norway
| | - Ragnhild A Lothe
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway.,Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Rolf I Skotheim
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway.,Department of Informatics, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
10
|
Berenguer CV, Pereira F, Câmara JS, Pereira JAM. Underlying Features of Prostate Cancer-Statistics, Risk Factors, and Emerging Methods for Its Diagnosis. Curr Oncol 2023; 30:2300-2321. [PMID: 36826139 PMCID: PMC9955741 DOI: 10.3390/curroncol30020178] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/09/2023] [Accepted: 02/12/2023] [Indexed: 02/17/2023] Open
Abstract
Prostate cancer (PCa) is the most frequently occurring type of malignant tumor and a leading cause of oncological death in men. PCa is very heterogeneous in terms of grade, phenotypes, and genetics, displaying complex features. This tumor often has indolent growth, not compromising the patient's quality of life, while its more aggressive forms can manifest rapid growth with progression to adjacent organs and spread to lymph nodes and bones. Nevertheless, the overtreatment of PCa patients leads to important physical, mental, and economic burdens, which can be avoided with careful monitoring. Early detection, even in the cases of locally advanced and metastatic tumors, provides a higher chance of cure, and patients can thus go through less aggressive treatments with fewer side effects. Furthermore, it is important to offer knowledge about how modifiable risk factors can be an effective method for reducing cancer risk. Innovations in PCa diagnostics and therapy are still required to overcome some of the limitations of the current screening techniques, in terms of specificity and sensitivity. In this context, this review provides a brief overview of PCa statistics, reporting its incidence and mortality rates worldwide, risk factors, and emerging screening strategies.
Collapse
Affiliation(s)
- Cristina V. Berenguer
- CQM—Centro de Química da Madeira, NPRG, Campus da Penteada, Universidade da Madeira, 9020-105 Funchal, Portugal
| | - Ferdinando Pereira
- SESARAM—Serviço de Saúde da Região Autónoma da Madeira, EPERAM, Hospital Dr. Nélio Mendonça, Avenida Luís de Camões 6180, 9000-177 Funchal, Portugal
| | - José S. Câmara
- CQM—Centro de Química da Madeira, NPRG, Campus da Penteada, Universidade da Madeira, 9020-105 Funchal, Portugal
- Departamento de Química, Faculdade de Ciências Exatas e Engenharia, Campus da Penteada, Universidade da Madeira, 9020-105 Funchal, Portugal
| | - Jorge A. M. Pereira
- CQM—Centro de Química da Madeira, NPRG, Campus da Penteada, Universidade da Madeira, 9020-105 Funchal, Portugal
- Correspondence:
| |
Collapse
|
11
|
Axcrona K, Aas K, Axcrona U, Skotheim RI. Re: Spatially Resolved Clonal Copy Number Alterations in Benign and Malignant Tissue. Eur Urol 2023; 83:183. [PMID: 36464529 DOI: 10.1016/j.eururo.2022.11.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 11/15/2022] [Indexed: 12/03/2022]
Affiliation(s)
- Karol Axcrona
- Department of Urology, Akershus University Hospital, Lørenskog, Norway; Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway.
| | - Kirsti Aas
- Department of Urology, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| | - Ulrika Axcrona
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway; Department of Pathology, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| | - Rolf I Skotheim
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway; Department of Informatics, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
12
|
Wong HY, Sheng Q, Hesterberg AB, Croessmann S, Rios BL, Giri K, Jackson J, Miranda AX, Watkins E, Schaffer KR, Donahue M, Winkler E, Penson DF, Smith JA, Herrell SD, Luckenbaugh AN, Barocas DA, Kim YJ, Graves D, Giannico GA, Rathmell JC, Park BH, Gordetsky JB, Hurley PJ. Single cell analysis of cribriform prostate cancer reveals cell intrinsic and tumor microenvironmental pathways of aggressive disease. Nat Commun 2022; 13:6036. [PMID: 36229464 PMCID: PMC9562361 DOI: 10.1038/s41467-022-33780-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 10/03/2022] [Indexed: 12/03/2022] Open
Abstract
Cribriform prostate cancer, found in both invasive cribriform carcinoma (ICC) and intraductal carcinoma (IDC), is an aggressive histological subtype that is associated with progression to lethal disease. To delineate the molecular and cellular underpinnings of ICC/IDC aggressiveness, this study examines paired ICC/IDC and benign prostate surgical samples by single-cell RNA-sequencing, TCR sequencing, and histology. ICC/IDC cancer cells express genes associated with metastasis and targets with potential for therapeutic intervention. Pathway analyses and ligand/receptor status model cellular interactions among ICC/IDC and the tumor microenvironment (TME) including JAG1/NOTCH. The ICC/IDC TME is hallmarked by increased angiogenesis and immunosuppressive fibroblasts (CTHRC1+ASPN+FAP+ENG+) along with fewer T cells, elevated T cell dysfunction, and increased C1QB+TREM2+APOE+-M2 macrophages. These findings support that cancer cell intrinsic pathways and a complex immunosuppressive TME contribute to the aggressive phenotype of ICC/IDC. These data highlight potential therapeutic opportunities to restore immune signaling in patients with ICC/IDC that may afford better outcomes.
Collapse
Affiliation(s)
- Hong Yuen Wong
- grid.412807.80000 0004 1936 9916Department of Medicine, Vanderbilt University Medical Center, Nashville, TN USA
| | - Quanhu Sheng
- grid.412807.80000 0004 1936 9916Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN USA
| | - Amanda B. Hesterberg
- grid.412807.80000 0004 1936 9916Department of Medicine, Vanderbilt University Medical Center, Nashville, TN USA
| | - Sarah Croessmann
- grid.412807.80000 0004 1936 9916Department of Medicine, Vanderbilt University Medical Center, Nashville, TN USA
| | - Brenda L. Rios
- grid.412807.80000 0004 1936 9916Department of Medicine, Vanderbilt University Medical Center, Nashville, TN USA
| | - Khem Giri
- grid.412807.80000 0004 1936 9916Department of Medicine, Vanderbilt University Medical Center, Nashville, TN USA
| | - Jorgen Jackson
- grid.412807.80000 0004 1936 9916Department of Medicine, Vanderbilt University Medical Center, Nashville, TN USA
| | - Adam X. Miranda
- grid.412807.80000 0004 1936 9916Department of Medicine, Vanderbilt University Medical Center, Nashville, TN USA
| | - Evan Watkins
- grid.412807.80000 0004 1936 9916Department of Medicine, Vanderbilt University Medical Center, Nashville, TN USA
| | - Kerry R. Schaffer
- grid.412807.80000 0004 1936 9916Department of Medicine, Vanderbilt University Medical Center, Nashville, TN USA ,grid.412807.80000 0004 1936 9916Vanderbilt-Ingram Cancer Center, Nashville, TN USA
| | - Meredith Donahue
- grid.412807.80000 0004 1936 9916Department of Urology, Vanderbilt University Medical Center, Nashville, TN USA
| | - Elizabeth Winkler
- grid.412807.80000 0004 1936 9916Department of Urology, Vanderbilt University Medical Center, Nashville, TN USA
| | - David F. Penson
- grid.412807.80000 0004 1936 9916Vanderbilt-Ingram Cancer Center, Nashville, TN USA ,grid.412807.80000 0004 1936 9916Department of Urology, Vanderbilt University Medical Center, Nashville, TN USA
| | - Joseph A. Smith
- grid.412807.80000 0004 1936 9916Department of Urology, Vanderbilt University Medical Center, Nashville, TN USA
| | - S. Duke Herrell
- grid.412807.80000 0004 1936 9916Department of Urology, Vanderbilt University Medical Center, Nashville, TN USA
| | - Amy N. Luckenbaugh
- grid.412807.80000 0004 1936 9916Department of Urology, Vanderbilt University Medical Center, Nashville, TN USA
| | - Daniel A. Barocas
- grid.412807.80000 0004 1936 9916Department of Urology, Vanderbilt University Medical Center, Nashville, TN USA
| | - Young J. Kim
- grid.412807.80000 0004 1936 9916Vanderbilt-Ingram Cancer Center, Nashville, TN USA ,grid.412807.80000 0004 1936 9916Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, TN USA ,grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals, Tarrytown, New York, USA
| | - Diana Graves
- grid.412807.80000 0004 1936 9916Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN USA
| | - Giovanna A. Giannico
- grid.412807.80000 0004 1936 9916Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN USA
| | - Jeffrey C. Rathmell
- grid.412807.80000 0004 1936 9916Vanderbilt-Ingram Cancer Center, Nashville, TN USA ,grid.412807.80000 0004 1936 9916Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN USA ,Vanderbilt Center for Immunobiology, Nashville, TN USA
| | - Ben H. Park
- grid.412807.80000 0004 1936 9916Department of Medicine, Vanderbilt University Medical Center, Nashville, TN USA ,grid.412807.80000 0004 1936 9916Vanderbilt-Ingram Cancer Center, Nashville, TN USA
| | - Jennifer B. Gordetsky
- grid.412807.80000 0004 1936 9916Vanderbilt-Ingram Cancer Center, Nashville, TN USA ,grid.412807.80000 0004 1936 9916Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN USA
| | - Paula J. Hurley
- grid.412807.80000 0004 1936 9916Department of Medicine, Vanderbilt University Medical Center, Nashville, TN USA ,grid.412807.80000 0004 1936 9916Vanderbilt-Ingram Cancer Center, Nashville, TN USA ,grid.412807.80000 0004 1936 9916Department of Urology, Vanderbilt University Medical Center, Nashville, TN USA
| |
Collapse
|
13
|
An C, Wang I, Li X, Xia R, Deng F. Long non-coding RNA in prostate cancer. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2022; 10:170-179. [PMID: 35874287 PMCID: PMC9301060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/15/2022] [Indexed: 06/15/2023]
Abstract
Prostate cancer is the most frequently diagnosed cancer in males and its development and progression remains an important area of study. Recently, long non-coding RNAs (lncRNAs) have been evidenced as key players in cancer pathogenesis. Specifically, dysregulation of long non-coding RNA (lncRNA) expression has shown to affect tumor proliferation and metastasis, acting as either tumor suppressors or oncogenes. However, its specific mechanisms and functions in prostate cancer remain unclear. This review provides an overview of currently available information on prostate cancer-related lncRNAs, including GAS5, GAS-007, MEG3, PCA3, PCAT14, PCAT1, PVT1, UCA1, SChLAP1, MALAT1, HOTAIR, and NEAT1. Notable tumor growth inhibitors include GAS5 and MEG3. GAS5 is evidenced to interfere with the AKT/MTOR signaling pathway through targeting microRNA mir-103. MEG3, however, is proposed to inhibit the cycle, sponge miR-9-5p, and induce gene silencing. PCAT1, PVT1, and UCA1 are important tumor growth promoters. PCAT1 is indicated to be a transcriptional repressor, a mir-145-5P sponge, and a P13K/AKT pathway activator. Studies suggest that PVT1 acts via microRNA targeting and regulating proliferating cell nuclear antigen. UCA1 may sponge miR-204 and miR-331-3p as well as regulate myosin VI. Thorough understanding of these lncRNAs may elucidate new aspects of prostate cancer pathology and serve a pivotal role in developing novel diagnostic and prognostic techniques.
Collapse
Affiliation(s)
- Christine An
- Institute of Human Nutrition, Columbia UniversityNew York, NY, USA
| | - Ian Wang
- State University of New York, Hofstra UniversityHempstead, NY, USA
| | - Xin Li
- Department of Molecular Pathobiology, New York University College of DentistryNY, USA
| | - Rong Xia
- Department of Pathology and Urology, New York University School of MedicineNY, USA
| | - Fangming Deng
- Department of Pathology and Urology, New York University School of MedicineNY, USA
| |
Collapse
|
14
|
Kidd SG, Bogaard M, Carm KT, Bakken AC, Maltau AMV, Løvf M, Lothe RA, Axcrona K, Axcrona U, Skotheim RI. In situ
expression of
ERG
protein in the context of tumor heterogeneity identifies prostate cancer patients with inferior prognosis. Mol Oncol 2022; 16:2810-2822. [PMID: 35574900 PMCID: PMC9348599 DOI: 10.1002/1878-0261.13225] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/29/2022] [Accepted: 05/13/2022] [Indexed: 11/12/2022] Open
Abstract
Prognostic biomarkers for prostate cancer are needed to improve prediction of disease course and guide treatment decisions. However, biomarker development is complicated by the common multifocality and heterogeneity of the disease. We aimed to determine the prognostic value of candidate biomarkers transcriptional regulator ERG and related ETS family genes, while considering tumor heterogeneity. In a multisampled, prospective, and treatment‐naïve radical prostatectomy cohort from one tertiary center (2010–2012, median follow‐up 8.1 years), we analyzed ERG protein (480 patients; 2047 tissue cores), and RNA of several ETS genes in a subcohort (165 patients; 778 fresh‐frozen tissue samples). Intra‐ and interfocal heterogeneity was identified in 29% and 33% (ERG protein) and 39% and 27% (ETS RNA) of patients, respectively. ERG protein and ETS RNA was identified exclusively in a nonindex tumor in 31% and 32% of patients, respectively. ERG protein demonstrated independent prognostic value in predicting biochemical (P = 0.04) and clinical recurrence (P = 0.004) and appeared to have greatest prognostic value for patients with Grade Groups 4–5. In conclusion, when heterogeneity is considered, ERG protein is a robust prognostic biomarker for prostate cancer.
Collapse
Affiliation(s)
- Susanne G. Kidd
- Department of Molecular Oncology, Institute for Cancer Research Oslo University Hospital–Radiumhospitalet Oslo Norway
- Institute for Clinical Medicine, Faculty of Medicine University of Oslo Oslo Norway
| | - Mari Bogaard
- Department of Molecular Oncology, Institute for Cancer Research Oslo University Hospital–Radiumhospitalet Oslo Norway
- Institute for Clinical Medicine, Faculty of Medicine University of Oslo Oslo Norway
- Department of Pathology Oslo University Hospital–Radiumhospitalet Oslo Norway
| | - Kristina T. Carm
- Department of Molecular Oncology, Institute for Cancer Research Oslo University Hospital–Radiumhospitalet Oslo Norway
| | - Anne Cathrine Bakken
- Department of Molecular Oncology, Institute for Cancer Research Oslo University Hospital–Radiumhospitalet Oslo Norway
| | - Aase M. V. Maltau
- Department of Molecular Oncology, Institute for Cancer Research Oslo University Hospital–Radiumhospitalet Oslo Norway
| | - Marthe Løvf
- Department of Molecular Oncology, Institute for Cancer Research Oslo University Hospital–Radiumhospitalet Oslo Norway
| | - Ragnhild A. Lothe
- Department of Molecular Oncology, Institute for Cancer Research Oslo University Hospital–Radiumhospitalet Oslo Norway
- Institute for Clinical Medicine, Faculty of Medicine University of Oslo Oslo Norway
| | - Karol Axcrona
- Department of Molecular Oncology, Institute for Cancer Research Oslo University Hospital–Radiumhospitalet Oslo Norway
- Department of Urology Akershus University Hospital Lørenskog Norway
| | - Ulrika Axcrona
- Department of Molecular Oncology, Institute for Cancer Research Oslo University Hospital–Radiumhospitalet Oslo Norway
- Department of Pathology Oslo University Hospital–Radiumhospitalet Oslo Norway
| | - Rolf I. Skotheim
- Department of Molecular Oncology, Institute for Cancer Research Oslo University Hospital–Radiumhospitalet Oslo Norway
- Department of Informatics, Faculty of Mathematics and Natural Sciences University of Oslo Oslo Norway
| |
Collapse
|
15
|
Pantazopoulos H, Diop MK, Grosset AA, Rouleau-Gagné F, Al-Saleh A, Boblea T, Trudel D. Intraductal Carcinoma of the Prostate as a Cause of Prostate Cancer Metastasis: A Molecular Portrait. Cancers (Basel) 2022; 14:820. [PMID: 35159086 PMCID: PMC8834356 DOI: 10.3390/cancers14030820] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 02/01/2023] Open
Abstract
Intraductal carcinoma of the prostate (IDC-P) is one of the most aggressive types of prostate cancer (PCa). IDC-P is identified in approximately 20% of PCa patients and is associated with recurrence, metastasis, and PCa-specific death. The main feature of this histological variant is the colonization of benign glands by PCa cells. Although IDC-P is a well-recognized independent parameter for metastasis, mechanisms by which IDC-P cells can spread and colonize other tissues are not fully known. In this review, we discuss the molecular portraits of IDC-P determined by immunohistochemistry and genomic approaches and highlight the areas in which more research is needed.
Collapse
Affiliation(s)
- Helen Pantazopoulos
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), 900 Saint-Denis, Montreal, QC H2X 0A9, Canada; (H.P.); (M.-K.D.); (A.-A.G.); (F.R.-G.); (A.A.-S.); (T.B.)
- Institut du Cancer de Montréal, 900 Saint-Denis, Montreal, QC H2X 0A9, Canada
- Department of Pathology and Cellular Biology, Université de Montréal, 2900 Boulevard Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
| | - Mame-Kany Diop
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), 900 Saint-Denis, Montreal, QC H2X 0A9, Canada; (H.P.); (M.-K.D.); (A.-A.G.); (F.R.-G.); (A.A.-S.); (T.B.)
- Institut du Cancer de Montréal, 900 Saint-Denis, Montreal, QC H2X 0A9, Canada
- Department of Pathology and Cellular Biology, Université de Montréal, 2900 Boulevard Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
| | - Andrée-Anne Grosset
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), 900 Saint-Denis, Montreal, QC H2X 0A9, Canada; (H.P.); (M.-K.D.); (A.-A.G.); (F.R.-G.); (A.A.-S.); (T.B.)
- Institut du Cancer de Montréal, 900 Saint-Denis, Montreal, QC H2X 0A9, Canada
- Department of Pathology and Cellular Biology, Université de Montréal, 2900 Boulevard Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
| | - Frédérique Rouleau-Gagné
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), 900 Saint-Denis, Montreal, QC H2X 0A9, Canada; (H.P.); (M.-K.D.); (A.-A.G.); (F.R.-G.); (A.A.-S.); (T.B.)
- Institut du Cancer de Montréal, 900 Saint-Denis, Montreal, QC H2X 0A9, Canada
- Department of Pathology and Cellular Biology, Université de Montréal, 2900 Boulevard Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
| | - Afnan Al-Saleh
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), 900 Saint-Denis, Montreal, QC H2X 0A9, Canada; (H.P.); (M.-K.D.); (A.-A.G.); (F.R.-G.); (A.A.-S.); (T.B.)
- Institut du Cancer de Montréal, 900 Saint-Denis, Montreal, QC H2X 0A9, Canada
- Department of Pathology and Cellular Biology, Université de Montréal, 2900 Boulevard Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
| | - Teodora Boblea
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), 900 Saint-Denis, Montreal, QC H2X 0A9, Canada; (H.P.); (M.-K.D.); (A.-A.G.); (F.R.-G.); (A.A.-S.); (T.B.)
- Institut du Cancer de Montréal, 900 Saint-Denis, Montreal, QC H2X 0A9, Canada
| | - Dominique Trudel
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), 900 Saint-Denis, Montreal, QC H2X 0A9, Canada; (H.P.); (M.-K.D.); (A.-A.G.); (F.R.-G.); (A.A.-S.); (T.B.)
- Institut du Cancer de Montréal, 900 Saint-Denis, Montreal, QC H2X 0A9, Canada
- Department of Pathology and Cellular Biology, Université de Montréal, 2900 Boulevard Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
- Department of Pathology, Centre Hospitalier de l’Université de Montréal (CHUM), 1051 Sanguinet, Montreal, QC H2X 0C1, Canada
| |
Collapse
|
16
|
Liu Q. The Emerging Landscape of Long Non-Coding RNAs in Wilms Tumor. Front Oncol 2022; 11:780925. [PMID: 35127486 PMCID: PMC8807488 DOI: 10.3389/fonc.2021.780925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/29/2021] [Indexed: 11/17/2022] Open
Abstract
Long noncoding RNAs (LncRNAs) are transcripts of nucleic acid sequences with a length of more than 200 bp, which have only partial coding capabilities. Recent studies have shown that lncRNAs located in the nucleus or cytoplasm can be used as gene expression regulatory elements due to their important regulatory effects in a variety of biological processes. Wilms tumor (WT) is a common abdominal tumor in children whose pathogenesis remains unclear. In recent years, many specifically expressed lncRNAs have been found in WT, which affect the occurrence and development of WT. At the same time, lncRNAs may have the capacity to become novel biomarkers for the diagnosis and prognosis of WT. This article reviews related research progress on the relationship between lncRNAs and WT, to provide a new direction for clinical diagnosis and treatment of WT.
Collapse
|
17
|
Skotheim RI, Axcrona U, Axcrona K. Re: Fibroblast Growth Factor Receptor 1 Drives the Metastatic Progression of Prostate Cancer. Eur Urol 2022; 81:431. [PMID: 35031159 DOI: 10.1016/j.eururo.2021.12.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 12/21/2021] [Indexed: 11/04/2022]
Affiliation(s)
- Rolf I Skotheim
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway; Department of Informatics, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Ulrika Axcrona
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway; Department of Pathology, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Karol Axcrona
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway; Department of Urology, Akershus University Hospital, Lørenskog, Norway.
| |
Collapse
|