1
|
Wei X, Zhao J, Nie L, Shi Y, Zhao F, Shen Y, Chen J, Sun G, Zhang X, Liang J, Hu X, Shen P, Chen N, Zeng H, Liu Z. Assessing the predictive value of intraductal carcinoma of the prostate (IDC-P) in determining abiraterone efficacy for metastatic hormone-sensitive prostate cancer (mHSPC) patients. Prostate 2024. [PMID: 39465570 DOI: 10.1002/pros.24809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/29/2024]
Abstract
BACKGROUND This study explored the value of intraductal carcinoma of the prostate (IDC-P) in predicting the efficacy of abiraterone treatment in metastatic hormone-sensitive prostate cancer (mHSPC) patients. METHODS A retrospective study of 925 patients who underwent prostate biopsies to detect IDC-P was conducted, with participants divided into two cohorts. The first cohort of 165 mHSPC patients receiving abiraterone treatment was analyzed to compare therapeutic effectiveness between IDC-P positive and negative cases. Utilizing propensity score matching (PSM) to reduce bias, outcomes such as PSA response, progression-free survival (PSA-PFS), radiographic progression-free survival (rPFS), and overall survival were assessed. Additionally, the second cohort of 760 mHSPC patients compared the efficacy of abiraterone with conventional hormone therapy, focusing on differences between IDC-P positive and negative individuals. RESULTS After PSM, our first cohort included 108 patients with similar baseline characteristics. Among them, 50% (54/108) were diagnosed with IDC-P, with 22.2% (12/54) having IDC-P pattern 1 and 77.8% (42/54) with IDC-P pattern 2. While no notable difference was seen in PSA responses between IDC-P positive and negative patients, IDC-P presence linked to worse clinical outcomes (PSA-PFS: 18.6 months vs. not reached [NR], p = 0.009; rPFS: 23.6 months vs. NR, p = 0.020). Further analysis showed comparable outcomes for IDC-P pattern 1 but significantly worse prognosis for IDC-P pattern 2 (PSA-PFS: 18.6 months vs. NR, p = 0.002; rPFS: 22.4 months vs. NR, p = 0.010). Subgroup analysis revealed IDC-P pattern 2 consistently predicted poorer outcomes across patient subgroups. Remarkably, both IDC-P positive and negative patients gained more from androgen deprivation therapy with abiraterone than conventional treatment, with IDC-P negative patients showing a more significant survival advantage, supported by better hazard ratios (0.47 and 0.66). CONCLUSION This study found that IDC-P, especially pattern 2, predicts poor prognosis in mHSPC patients on abiraterone therapy. Also, abiraterone's advantage over hormone therapy is reduced in cases with IDC-P compared to those without.
Collapse
Affiliation(s)
- Xinyuan Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Jinge Zhao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Ling Nie
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Yifu Shi
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Fengnian Zhao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Shen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Junru Chen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Guangxi Sun
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Xingming Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Jiayu Liang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Xu Hu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Pengfei Shen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Ni Chen
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Zeng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Zhenhua Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
2
|
Huang H, Zou S, Wan J, Zeng X, Wang S, Hu Z, Zhu X, Yang C. 68Ga-PSMA PET/CT and 18F-FDG PET/CT in the diagnosis of prostatic ductal cancer. Urol Oncol 2024:S1078-1439(24)00564-7. [PMID: 39098476 DOI: 10.1016/j.urolonc.2024.07.011] [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: 03/14/2024] [Revised: 06/30/2024] [Accepted: 07/15/2024] [Indexed: 08/06/2024]
Abstract
PURPOSES To explore the characteristics of PSMA PET/CT and FDG PET/CT images in prostatic ductal adenocarcinoma (DA) patients. METHODS We retrospectively enrolled prostatic DA patients with PET/CT scans at Tongji Hospital from 2018 to 2022. Patients with prostatic acinar adenocarcinoma (AA) and benign pathology (BP) were enrolled by 1:1 matching. Differences in the uptake of primary and metastatic foci on PET among the groups were analyzed. RESULTS A total of 42 patients were enrolled: 14 in each group. In primary foci, the mean PSMA uptake in the DA group was lower than that in the AA group (14.2 ± 9.6 vs. 27.1 ± 14.3, P = 0.009) and greater than that in the BP group (14.2 ± 9.6 vs. 4.7 ± 1.3, P = 0.003). The AUCs of the DA-AA ROC curve and DA-BP ROC curve were 0.781 and 0.872, respectively. The median PSMA uptake of metastatic lymph nodes in the DA group was lower than that in the AA group (5.6 vs. 14.2, P = 0.033), with no significant difference in metastatic bone lesions (9.5 vs 19.1, P = 0.485). No significant difference was found in the FDG uptake of primary and metastatic foci between the DA and AA groups (P > 0.05). CONCLUSION Prostatic DA has greater PSMA uptake than BP diseases, but lower uptake in both primary foci and metastatic lymph nodes than AA on PSMA PET/CT, aiding in the differential diagnosis of DA, AA and BP diseases. Clinicians should combine traditional imaging with PSMA PET/CT to avoid underestimating the clinical stage of DA patients.
Collapse
Affiliation(s)
- Haijun Huang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sijuan Zou
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Wan
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Zeng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shaogang Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiquan Hu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiaohua Zhu
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Chunguang Yang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
3
|
Cui Y, Lin J, Sun D, Zhang H, Diao T, Fu Q. Nomogram for predicting the overall survival and cancer-specific survival of patients with intraductal carcinoma of the prostate. J Cancer Res Clin Oncol 2024; 150:45. [PMID: 38281261 PMCID: PMC10822789 DOI: 10.1007/s00432-023-05582-5] [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: 11/09/2023] [Accepted: 11/21/2023] [Indexed: 01/30/2024]
Abstract
PURPOSE Intraductal carcinoma of the prostate (IDC-P) is a histological subtype that differs from conventional acinar adenocarcinoma in terms of its origin, appearance, and pathological features. For IDC-P, there is currently no recognized best course of action, and its prognosis is unclear. The goal of this study is to analyze independent prognostic factors in IDC-P patients and to develop and validate a nomogram to predict overall survival (OS) and cancer-specific survival (CSS). METHODS Clinical data for IDC-P patients were collected from the Surveillance, Epidemiology, and End Results database. To identify the independent variables influencing prognosis, multivariate Cox regression analysis was performed. A nomogram model was created utilizing these variables after comparing the variations in OS and CSS among various subgroups using Kaplan‒Meier curves. Internal validation of the nomograms was verified using the bootstrap resampling method. RESULTS The study included 280 IDC-P patients in total. Marital status, summary stage, grade, and the presence of lung metastases were significant factors impacting OS, and CSS was significantly influenced by marital status, summary stage, AJCC stage, the presence of lung metastases, the presence of bone metastases, and PSA according to univariate and multivariate Cox regression models (P < 0.05). Nomogram models were created to estimate OS and CSS using these parameters. The OS prediction model's C-index was 0.744, whereas the CSS prediction model's C-index was 0.831. CONCLUSION We developed and verified nomogram models for the prediction of 1-, 3-, and 5-year OS and CSS in patients with IDC-P. These nomograms serve as a resource for evaluating patient prognosis, therapy, and diagnosis, ultimately improving clinical decision-making accuracy.
Collapse
Affiliation(s)
- Yongqiang Cui
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Rd, Jinan, 250021, Shandong, China
| | - Junyang Lin
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Rd, Jinan, 250021, Shandong, China
| | - Dingqi Sun
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Rd, Jinan, 250021, Shandong, China
| | - Hui Zhang
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Rd, Jinan, 250021, Shandong, China
| | - Tongxiang Diao
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Rd, Jinan, 250021, Shandong, China.
| | - Qiang Fu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Rd, Jinan, 250021, Shandong, China.
- Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, China.
- Key Laboratory of Urinary Diseases in Universities of Shandong Shandong First Medical University, Jinan, 250021, China.
| |
Collapse
|
4
|
Naito Y, Kato M, Nagayama J, Sano Y, Matsuo K, Inoue S, Sano T, Ishida S, Matsukawa Y, Tsuzuki T, Akamatsu S. Recent insights on the clinical, pathological, and molecular features of intraductal carcinoma of the prostate. Int J Urol 2024; 31:7-16. [PMID: 37728330 DOI: 10.1111/iju.15299] [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: 06/26/2023] [Accepted: 09/06/2023] [Indexed: 09/21/2023]
Abstract
Intraductal carcinoma of the prostate, a unique histopathologic entity that is often observed (especially in advanced prostate cancer), is characterized by the proliferation of malignant cells within normal acini or ducts surrounded by a basement membrane. Intraductal carcinoma of the prostate is almost invariably associated with an adjacent high-grade carcinoma and is occasionally observed as an isolated subtype. Intraductal carcinoma of the prostate has been demonstrated to be an independent poor prognostic factor for all stages of cancer, whether localized, de novo metastatic, or castration-resistant. It also has a characteristic genetic profile, including high genomic instability. Recognizing and differentiating it from other pathologies is therefore important in patient management, and morphological diagnostic criteria for intraductal carcinoma of the prostate have been established. This review summarizes and outlines the clinical and pathological features, differential diagnosis, molecular aspects, and management of intraductal carcinoma of the prostate, as described in previous studies. We also present a discussion and future perspectives regarding intraductal carcinoma of the prostate.
Collapse
Affiliation(s)
- Yushi Naito
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Masashi Kato
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Jun Nagayama
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Yuta Sano
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Kazuna Matsuo
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Satoshi Inoue
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Tomoyasu Sano
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Shohei Ishida
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Yoshihisa Matsukawa
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Toyonori Tsuzuki
- Department of Surgical Pathology, Aichi Medical University Hospital, Nagakute, Aichi, Japan
| | - Shusuke Akamatsu
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| |
Collapse
|
5
|
Zhu S, Xu N, Zeng H. Molecular complexity of intraductal carcinoma of the prostate. Cancer Med 2024; 13:e6939. [PMID: 38379333 PMCID: PMC10879723 DOI: 10.1002/cam4.6939] [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: 06/27/2023] [Revised: 11/21/2023] [Accepted: 12/04/2023] [Indexed: 02/22/2024] Open
Abstract
Intraductal carcinoma of the prostate (IDC-P) is an aggressive subtype of prostate cancer characterized by the growth of tumor cells within the prostate ducts. It is often found alongside invasive carcinoma and is associated with poor prognosis. Understanding the molecular mechanisms driving IDC-P is crucial for improved diagnosis, prognosis, and treatment strategies. This review summarizes the molecular characteristics of IDC-P and their prognostic indications, comparing them to conventional prostate acinar adenocarcinoma, to gain insights into its unique behavior and identify potential therapeutic targets.
Collapse
Affiliation(s)
- Sha Zhu
- Department of Urology, Institute of Urology, West China HospitalSichuan UniversityChengduChina
| | - Nanwei Xu
- Department of Urology, Institute of Urology, West China HospitalSichuan UniversityChengduChina
| | - Hao Zeng
- Department of Urology, Institute of Urology, West China HospitalSichuan UniversityChengduChina
| |
Collapse
|
6
|
Wang X, Zhou L, Qi L, Zhang Y, Yin H, Gan Y, Gao X, Cai Y. High GLUT1 membrane expression and low PSMA membrane expression in Ductal Adenocarcinoma and Intraductal Carcinoma of the prostate. Prostate Cancer Prostatic Dis 2023:10.1038/s41391-023-00759-y. [PMID: 38007533 DOI: 10.1038/s41391-023-00759-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/02/2023] [Accepted: 11/14/2023] [Indexed: 11/27/2023]
Abstract
BACKGROUND Both Ductal Adenocarcinoma (DAC) and Intraductal Carcinoma (IDC) of the prostate are generally associated with aggressive clinical behavior and poor prognosis, which were linked with discordant FDG positivity and low Prostate-Specific Membrane Antigen (PSMA) expression. A recent study only cited a DAC patient with low 68Ga-PSMA-11 PET/CT uptake but high 18F-FDG PET/CT uptake, however, there is lack of directly compared articles nor large data sets. Hence, the objective of this study was to investigate the expression of PSMA and GLUT1 in DAC and IDC-P patients. METHODS The study was conducted on 87 DAC or/and IDC-P patients without any treatment and 97 PAC patients with a Gleason score ≥8 of prostate biopsies and prostatectomy samples between August 2017 and August 2022. We performed immunohistochemical staining and scoring of various cancer component samples from the patients to reflect the protein expression levels of PSMA and GLUT1. RESULTS PSMA expression in PAC was significantly higher than in DAC/IDC-P (141.2 vs 78.6, p < 0.001). There was no significant difference in PSMA expression between DAC/IDC-P and adjacent PAC (78.6 vs 93.4, p = 0.166). GLUT1 expression was higher in DAC/IDC-P than in adjacent PAC (68.6 vs 51.3, p = 0.007), but was still lower than that in pure PAC (68.6 vs 93.1, p = 0.0014). It is worth noting that GLUT1 membrane expression in DAC/IDC-P was significantly increased than in pure PAC (13.0 vs 6.6, p = 0.025), and in PAC adjacent to DAC/IDC-P (13.0 vs 2.0, p < 0.001). CONCLUSIONS In DAC/IDC-P tissues, PSMA expression is low, while GLUT1 expression, especially GLUT1 membrane expression is high. These findings imply that DAC/IDC-P may have higher glucose metabolic and raise interest in targeting membrane GLUT1 as a novel anticancer strategy for DAC/IDC-P and other prostate cancer with high glucose metabolism.
Collapse
Affiliation(s)
- Xingming Wang
- Department of Urology, Disorders of Prostate Cancer Multidisciplinary Team, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Li Zhou
- Department of Pathology, Disorders of Prostate Cancer Multidisciplinary Team, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Qi
- Department of Urology, Disorders of Prostate Cancer Multidisciplinary Team, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ye Zhang
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Disorders of Prostate Cancer Multidisciplinary Team, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hongling Yin
- Department of Pathology, Disorders of Prostate Cancer Multidisciplinary Team, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yu Gan
- Department of Urology, Disorders of Prostate Cancer Multidisciplinary Team, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Xiaomei Gao
- Department of Pathology, Disorders of Prostate Cancer Multidisciplinary Team, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Yi Cai
- Department of Urology, Disorders of Prostate Cancer Multidisciplinary Team, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
7
|
Lawrence MG, Taylor RA, Cuffe GB, Ang LS, Clark AK, Goode DL, Porter LH, Le Magnen C, Navone NM, Schalken JA, Wang Y, van Weerden WM, Corey E, Isaacs JT, Nelson PS, Risbridger GP. The future of patient-derived xenografts in prostate cancer research. Nat Rev Urol 2023; 20:371-384. [PMID: 36650259 PMCID: PMC10789487 DOI: 10.1038/s41585-022-00706-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] [Accepted: 12/09/2022] [Indexed: 01/19/2023]
Abstract
Patient-derived xenografts (PDXs) are generated by engrafting human tumours into mice. Serially transplantable PDXs are used to study tumour biology and test therapeutics, linking the laboratory to the clinic. Although few prostate cancer PDXs are available in large repositories, over 330 prostate cancer PDXs have been established, spanning broad clinical stages, genotypes and phenotypes. Nevertheless, more PDXs are needed to reflect patient diversity, and to study new treatments and emerging mechanisms of resistance. We can maximize the use of PDXs by exchanging models and datasets, and by depositing PDXs into biorepositories, but we must address the impediments to accessing PDXs, such as institutional, ethical and legal agreements. Through collaboration, researchers will gain greater access to PDXs representing diverse features of prostate cancer.
Collapse
Affiliation(s)
- Mitchell G Lawrence
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia.
- Melbourne Urological Research Alliance, Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia.
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.
- Cabrini Institute, Cabrini Health, Malvern, Victoria, Australia.
| | - Renea A Taylor
- Melbourne Urological Research Alliance, Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Cabrini Institute, Cabrini Health, Malvern, Victoria, Australia
- Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Georgia B Cuffe
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Lisa S Ang
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Ashlee K Clark
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
- Department of Urology, Radboud University Medical Center, Nijmegen, Netherlands
| | - David L Goode
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Laura H Porter
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Clémentine Le Magnen
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
- Department of Urology, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Nora M Navone
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jack A Schalken
- Department of Urology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Yuzhuo Wang
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, British Columbia, Canada
| | | | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA, USA
| | - John T Isaacs
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter S Nelson
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Gail P Risbridger
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia.
- Melbourne Urological Research Alliance, Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia.
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.
- Cabrini Institute, Cabrini Health, Malvern, Victoria, Australia.
| |
Collapse
|
8
|
Roberts MJ, Maurer T, Perera M, Eiber M, Hope TA, Ost P, Siva S, Hofman MS, Murphy DG, Emmett L, Fendler WP. Using PSMA imaging for prognostication in localized and advanced prostate cancer. Nat Rev Urol 2023; 20:23-47. [PMID: 36473945 DOI: 10.1038/s41585-022-00670-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2022] [Indexed: 12/12/2022]
Abstract
The use of prostate-specific membrane antigen (PSMA)-directed applications in modern prostate cancer management has evolved rapidly over the past few years, helping to establish new treatment pathways and provide further insights into prostate cancer biology. However, the prognostic implications of PSMA-PET have not been studied systematically, owing to rapid clinical implementation without long follow-up periods to determine intermediate-term and long-term oncological outcomes. Currently available data suggest that traditional prognostic factors and survival outcomes are associated with high PSMA expression (both according to immunohistochemistry and PET uptake) in men with localized and biochemically recurrent disease. Treatment with curative intent (primary and/or salvage) often fails when PSMA-positive metastases are present; however, the sensitivity of PSMA-PET in detecting all metastases is poor. Low PSMA-PET uptake in recurrent disease is a favourable prognostic factor; however, it can be associated with poor prognosis in conjunction with high 18F-fluorodeoxyglucose uptake in metastatic castration-resistant prostate cancer. Clinical trials embedding PSMA-PET for guiding management with reliable oncological outcomes are needed to support ongoing clinical use.
Collapse
Affiliation(s)
- Matthew J Roberts
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia.
- University of Queensland Centre for Clinical Research, Faculty of Medicine, Brisbane, Queensland, Australia.
- Department of Urology, Redcliffe Hospital, Brisbane, Queensland, Australia.
| | - Tobias Maurer
- Martini-Klinik Prostate Cancer Center, Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Marlon Perera
- Department of Surgery, Austin Health, Heidelberg, Victoria, Australia
| | - Matthias Eiber
- Department of Nuclear Medicine, Technical University of Munich, Munich, Germany
| | - Thomas A Hope
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Piet Ost
- Department of Radiation Oncology, Iridium Network, GZA Ziekenhuizen, Antwerp, Belgium
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Shankar Siva
- Peter MacCallum Cancer Centre, Radiation Oncology, Parkville, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, Melbourne University, Parkville, Victoria, Australia
| | - Michael S Hofman
- Sir Peter MacCallum Department of Oncology, Melbourne University, Parkville, Victoria, Australia
- Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Declan G Murphy
- Sir Peter MacCallum Department of Oncology, Melbourne University, Parkville, Victoria, Australia
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Louise Emmett
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, New South Wales, Australia
- Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University of Duisburg-Essen, Essen, Germany
- PET Committee of the German Society of Nuclear Medicine, Goettingen, Germany
| |
Collapse
|
9
|
Homologous recombination deficiency (HRD) score in aggressive prostatic adenocarcinoma with or without intraductal carcinoma of the prostate (IDC-P). BMC Med 2022; 20:237. [PMID: 35864546 PMCID: PMC9306093 DOI: 10.1186/s12916-022-02430-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 06/07/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Intraductal carcinoma of the prostate (IDC-P) is a subtype of prostate cancer featured by poor prognosis. Previous studies suggested IDC-P could have a potentially unstable genome. Homologous recombination deficiency (HRD) score is a result-oriented method to describe the genomic instability status. This study investigates the association of HRD scores with IDC-P and other clinicopathological factors and the prognostic implication of HRD scores in an aggressive prostate cancer cohort. METHODS This study involved 123 PCa patients, including high-risk localized (M0) and de novo metastatic (M1) diseases. HRD score is calculated based on over 10,000 single-nucleotide polymorphisms distributed across the human genome. We explored the association between HRD scores and clinicopathological characteristics, genomic alterations, and patients' prognoses using rank-sum tests, chi-square tests, Kaplan-Meier curves, and Cox proportional hazards method. RESULTS The median HRD score of this cohort is 21.0, with 65 (52.8%) patients showing HRD score≥21. Tumors with IDC-P displayed higher HRD scores than adenocarcinoma (P=0.002); other high HRD score-related factors included M1 (P =0.008) and high ISUP grades (4-5) (P=0.001). MYC mutations were associated with high HRD scores (P<0.001) in the total cohort. TP53 mutations (P=0.010) and HRR pathway mutations (P=0.028) corresponded to high HRD scores in IDC-P positive and non-IDC-P patients, respectively, but not vice versa. HRD scores higher than 21 indicated significantly worse survival in the total cohort. CONCLUSIONS M1, high Gleason score, and IDC-P pathology represent higher HRD scores in PCa. Tumors with IDC-P might have different driven mechanisms for high HRD scores than non-IDC-P. HRD score displayed prognostic value in this aggressive prostate cancer cohort.
Collapse
|
10
|
Gillessen S, Armstrong A, Attard G, Beer TM, Beltran H, Bjartell A, Bossi A, Briganti A, Bristow RG, Bulbul M, Caffo O, Chi KN, Clarke CS, Clarke N, Davis ID, de Bono JS, Duran I, Eeles R, Efstathiou E, Efstathiou J, Ekeke ON, Evans CP, Fanti S, Feng FY, Fizazi K, Frydenberg M, George D, Gleave M, Halabi S, Heinrich D, Higano C, Hofman MS, Hussain M, James N, Jones R, Kanesvaran R, Khauli RB, Klotz L, Leibowitz R, Logothetis C, Maluf F, Millman R, Morgans AK, Morris MJ, Mottet N, Mrabti H, Murphy DG, Murthy V, Oh WK, Ost P, O'Sullivan JM, Padhani AR, Parker C, Poon DMC, Pritchard CC, Rabah DM, Rathkopf D, Reiter RE, Rubin M, Ryan CJ, Saad F, Sade JP, Sartor O, Scher HI, Shore N, Skoneczna I, Small E, Smith M, Soule H, Spratt DE, Sternberg CN, Suzuki H, Sweeney C, Sydes MR, Taplin ME, Tilki D, Tombal B, Türkeri L, Uemura H, Uemura H, van Oort I, Yamoah K, Ye D, Zapatero A, Omlin A. Management of Patients with Advanced Prostate Cancer: Report from the Advanced Prostate Cancer Consensus Conference 2021. Eur Urol 2022; 82:115-141. [PMID: 35450732 DOI: 10.1016/j.eururo.2022.04.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/01/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Innovations in treatments, imaging, and molecular characterisation in advanced prostate cancer have improved outcomes, but various areas of management still lack high-level evidence to inform clinical practice. The 2021 Advanced Prostate Cancer Consensus Conference (APCCC) addressed some of these questions to supplement guidelines that are based on level 1 evidence. OBJECTIVE To present the voting results from APCCC 2021. DESIGN, SETTING, AND PARTICIPANTS The experts identified three major areas of controversy related to management of advanced prostate cancer: newly diagnosed metastatic hormone-sensitive prostate cancer (mHSPC), the use of prostate-specific membrane antigen ligands in diagnostics and therapy, and molecular characterisation of tissue and blood. A panel of 86 international prostate cancer experts developed the programme and the consensus questions. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The panel voted publicly but anonymously on 107 pre-defined questions, which were developed by both voting and non-voting panel members prior to the conference following a modified Delphi process. RESULTS AND LIMITATIONS The voting reflected the opinions of panellists and did not incorporate a standard literature review or formal meta-analysis. The answer options for the consensus questions received varying degrees of support from panellists, as reflected in this article and the detailed voting results reported in the Supplementary material. CONCLUSIONS These voting results from a panel of experts in advanced prostate cancer can help clinicians and patients to navigate controversial areas of management for which high-level evidence is scant. However, diagnostic and treatment decisions should always be individualised according to patient characteristics, such as the extent and location of disease, prior treatment(s), comorbidities, patient preferences, and treatment recommendations, and should also incorporate current and emerging clinical evidence and logistic and economic constraints. Enrolment in clinical trials should be strongly encouraged. Importantly, APCCC 2021 once again identified salient questions that merit evaluation in specifically designed trials. PATIENT SUMMARY The Advanced Prostate Cancer Consensus Conference is a forum for discussing current diagnosis and treatment options for patients with advanced prostate cancer. An expert panel votes on predefined questions focused on the most clinically relevant areas for treatment of advanced prostate cancer for which there are gaps in knowledge. The voting results provide a practical guide to help clinicians in discussing treatment options with patients as part of shared decision-making.
Collapse
Affiliation(s)
- Silke Gillessen
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland; Universita della Svizzera Italiana, Lugano, Switzerland; University of Berne, Berne, Switzerland; Division of Cancer Sciences, University of Manchester, Manchester, UK.
| | - Andrew Armstrong
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, USA
| | - Gert Attard
- University College London Cancer Institute, London, UK
| | - Tomasz M Beer
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Himisha Beltran
- Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Anders Bjartell
- Department of Urology, Skåne University Hospital, Malmö, Sweden
| | - Alberto Bossi
- Genitourinary Oncology, Prostate Brachytherapy Unit, Gustave Roussy, Paris, France
| | - Alberto Briganti
- Unit of Urology/Division of Oncology, Urological Research Institute, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, Milan, Italy
| | - Robert G Bristow
- Division of Cancer Sciences, University of Manchester, Manchester, UK; Christie NHS Trust and CRUK Manchester Institute and Cancer Centre, Manchester, UK
| | - Muhammad Bulbul
- Division of Urology, Department of Surgery, American University of Beirut Medical Center, Beirut, Lebanon
| | - Orazio Caffo
- Department of Medical Oncology, Santa Chiara Hospital, Trento, Italy
| | - Kim N Chi
- BC Cancer, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Caroline S Clarke
- Research Department of Primary Care & Population Health, Royal Free Campus, University College London, London, UK
| | - Noel Clarke
- The Christie and Salford Royal Hospitals, Manchester, UK
| | - Ian D Davis
- Monash University and Eastern Health, Victoria, Australia
| | - Johann S de Bono
- The Institute of Cancer Research, Royal Marsden NHS Foundation Trust, Sutton, UK
| | - Ignacio Duran
- Department of Medical Oncology, Hospital Universitario Marques de Valdecilla, IDIVAL, Santander, Spain
| | - Ros Eeles
- The Institute of Cancer Research, Royal Marsden NHS Foundation Trust, Sutton, UK
| | | | - Jason Efstathiou
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Onyeanunam Ngozi Ekeke
- Department of Surgery, University of Port Harcourt Teaching Hospital, Port Harcourt, Nigeria
| | | | - Stefano Fanti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Felix Y Feng
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California-San Francisco, San Francisco, CA, USA
| | - Karim Fizazi
- Institut Gustave Roussy, University of Paris Saclay, Villejuif, France
| | - Mark Frydenberg
- Department of Surgery, Prostate Cancer Research Program, Monash University, Melbourne, Australia
| | - Dan George
- Departments of Medicine and Surgery, Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Martin Gleave
- Urological Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Susan Halabi
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Daniel Heinrich
- Department of Oncology and Radiotherapy, Innlandet Hospital Trust, Gjøvik, Norway
| | | | - Michael S Hofman
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Department of Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Maha Hussain
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Nick James
- The Institute of Cancer Research, Royal Marsden NHS Foundation Trust, Sutton, UK
| | - Robert Jones
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | | | - Raja B Khauli
- Department of Urology and the Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Beirut, Lebanon
| | - Laurence Klotz
- Division of Urology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Raya Leibowitz
- Oncology Institute, Shamir Medical Center and Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Chris Logothetis
- Department of Genitourinary Medical Oncology, David H. Koch Centre, MD Anderson Cancer Centre, Houston, TX, USA; Department of Clinical Therapeutics, University of Athens Alexandra Hospital, Athens, Greece
| | - Fernando Maluf
- Beneficiência Portuguesa de São Paulo, São Paulo, SP, Brazil; Departamento de Oncologia, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | | | - Alicia K Morgans
- Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael J Morris
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Hind Mrabti
- National Institute of Oncology, Mohamed V University, Rabat, Morocco
| | - Declan G Murphy
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | | | - William K Oh
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, The Tisch Cancer Institute, New York, NY, USA
| | - Piet Ost
- Department of Radiation Oncology, Iridium Netwerk, Antwerp, Belgium; Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Joe M O'Sullivan
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Northern Ireland Cancer Centre, Belfast City Hospital, Belfast, UK
| | - Anwar R Padhani
- The Institute of Cancer Research, Royal Marsden NHS Foundation Trust, Sutton, UK; Mount Vernon Cancer Centre, London, UK
| | - Chris Parker
- The Institute of Cancer Research, Royal Marsden NHS Foundation Trust, Sutton, UK
| | - Darren M C Poon
- Comprehensive Oncology Centre, Hong Kong Sanatorium & Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Colin C Pritchard
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Danny M Rabah
- The Cancer Research Chair, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Dana Rathkopf
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rob E Reiter
- University of California-Los Angeles, Los Angeles, CA, USA
| | - Mark Rubin
- Bern Center for Precision Medicine and Department for Biomedical Research, Bern, Switzerland
| | - Charles J Ryan
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Fred Saad
- Centre Hospitalier de Université de Montréal, Montreal, Canada
| | - Juan P Sade
- Instituto Alexander Fleming, Buenos Aires, Argentina
| | | | - Howard I Scher
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Neal Shore
- Carolina Urologic Research Center, Myrtle Beach, SC, USA
| | - Iwona Skoneczna
- Rafal Masztak Grochowski Hospital and Maria Sklodowska Curie National Research Institute of Oncology, Warsaw, Poland
| | - Eric Small
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California-San Francisco, San Francisco, CA, USA
| | - Matthew Smith
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Howard Soule
- Prostate Cancer Foundation, Santa Monica, CA, USA
| | - Daniel E Spratt
- University Hospitals Seidman Cancer Center, Cleveland, OH, USA
| | - Cora N Sternberg
- Englander Institute for Precision Medicine, Weill Cornell Medicine Division of Hematology and Oncology, Meyer Cancer Center, New York Presbyterian Hospital, New York, NY, USA
| | | | - Christopher Sweeney
- Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Matthew R Sydes
- MRC Clinical Trials Unit, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Mary-Ellen Taplin
- Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Derya Tilki
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Urology, Koc University Hospital, Istanbul, Turkey
| | | | - Levent Türkeri
- Department of Urology, M.A. Aydınlar Acıbadem University, Altunizade Hospital, Istanbul, Turkey
| | - Hiroji Uemura
- Yokohama City University Medical Center, Yokohama, Japan
| | - Hirotsugu Uemura
- Department of Urology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Inge van Oort
- Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kosj Yamoah
- Department of Radiation Oncology & Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, University of South Florida, Tampa, FL, USA
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Almudena Zapatero
- Department of Radiation Oncology, Hospital Universitario de La Princesa, Health Research Institute, Madrid, Spain
| | - Aurelius Omlin
- Department of Medical Oncology and Haematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| |
Collapse
|
11
|
Yang L, Li Z, Liang X, Xu J, Cai Y, Huang C, Zhang M, Yao J, Song B. Radiomic Machine Learning and External Validation Based on 3.0 T mpMRI for Prediction of Intraductal Carcinoma of Prostate With Different Proportion. Front Oncol 2022; 12:934291. [PMID: 35837116 PMCID: PMC9274129 DOI: 10.3389/fonc.2022.934291] [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: 05/02/2022] [Accepted: 05/24/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose To assess the association of radiomics features based on multiparametric MRI (mpMRI) with the proportion of intraductal carcinoma of prostate (IDC-P) and validate the predictive models. Materials and Methods We retrospectively included pre-treatment MR images of prostate cancer (PCa) with IDC components of high proportion (≥10%, hpIDC-P), low proportion (<10%, lpIDC-P), and pure acinar adenocarcinoma (PAC) from our institution for training and internal validation and cooperated cohort for external validation. Normalized images of T2WI, diffusion weighted imaging (DWI) and apparent diffusion coefficient (ADC) map, and dynamic contrast enhanced (DCE) sequences were used for radiomics modeling. The clinical model was built based on serum total prostate specific antigen (tPSA) and Gleason score (GS), and the integrated model was the combination of Rad-score and clinicopathological data. The discrimination ability was assessed by area under the receiver operating characteristic curve (ROC-AUC) in the internal and external validation sets and compared by DeLong test. Results Overall, 97 patients with hpIDC-P, 87 lpIDC-P, and 78 PAC were included for training and internal validation, and 11, 16, and 19 patients for external validation. The integrated model for predicting hpIDC-P got the best ROC-AUC of 0.88 (95%CI = 0.83-0.93) in internal and 0.86 (95%CI = 0.72-1.0) in external validation, which both outperformed clinical models (AUC=0.78, 95% CI = 0.72-0.85, AUC=0.69, 95% CI = 0.5-0.85, respectively) based solely on GS, and the radiomics model (AUC=0.85, 95% CI = 0.79-0.91) was slightly inferior to the integrated model and better than the clinical model in internal dataset. The integrated model for predicting lpIDC-P outperformed both radiomics and clinical models in the internal dataset, while slightly inferior to the integrated model for predicting hpIDC-P. Conclusions Radiomics signature improved differentiation of both hpIDC-P and lpIDC-P versus PAC when compared with the clinical model based on Gleason score, and was validated in an external cohort.
Collapse
Affiliation(s)
- Ling Yang
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Zhengyan Li
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Xu Liang
- Department of Radiology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jingxu Xu
- Department of Research Collaboration, R&D center, Beijing Deepwise & League of PHD Technology Co., Ltd., Beijing, China
| | - Yusen Cai
- Department of Research Collaboration, R&D center, Beijing Deepwise & League of PHD Technology Co., Ltd., Beijing, China
| | - Chencui Huang
- Department of Research Collaboration, R&D center, Beijing Deepwise & League of PHD Technology Co., Ltd., Beijing, China
| | - Mengni Zhang
- Department of Pathology, West China Hospital of Sichuan University, Chengdu, China
| | - Jin Yao
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Bin Song
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
12
|
Mai CW, Chin KY, Foong LC, Pang KL, Yu B, Shu Y, Chen S, Cheong SK, Chua CW. Modeling prostate cancer: What does it take to build an ideal tumor model? Cancer Lett 2022; 543:215794. [PMID: 35718268 DOI: 10.1016/j.canlet.2022.215794] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 06/10/2022] [Indexed: 11/17/2022]
Abstract
Prostate cancer is frequently characterized as a multifocal disease with great intratumoral heterogeneity as well as a high propensity to metastasize to bone. Consequently, modeling prostate tumor has remained a challenging task for researchers in this field. In the past decades, genomic advances have led to the identification of key molecular alterations in prostate cancer. Moreover, resistance towards second-generation androgen-deprivation therapy, namely abiraterone and enzalutamide has unveiled androgen receptor-independent diseases with distinctive histopathological and clinical features. In this review, we have critically evaluated the commonly used preclinical models of prostate cancer with respect to their capability of recapitulating the key genomic alterations, histopathological features and bone metastatic potential of human prostate tumors. In addition, we have also discussed the potential use of the emerging organoid models in prostate cancer research, which possess clear advantages over the commonly used preclinical tumor models. We anticipate that no single model can faithfully recapitulate the complexity of prostate cancer, and thus, propose the use of a cost- and time-efficient integrated tumor modeling approach for future prostate cancer investigations.
Collapse
Affiliation(s)
- Chun-Wai Mai
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China; Centre for Stem Cell Research, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Selangor, 43000, Malaysia
| | - Kok-Yong Chin
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China; Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, 56000, Malaysia
| | - Lian-Chee Foong
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China; Centre for Stem Cell Research, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Selangor, 43000, Malaysia
| | - Kok-Lun Pang
- Newcastle University Medicine Malaysia, Iskandar Puteri, 79200, Malaysia
| | - Bin Yu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yu Shu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Sisi Chen
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Soon-Keng Cheong
- Centre for Stem Cell Research, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Selangor, 43000, Malaysia
| | - Chee Wai Chua
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
13
|
Tryggestad AMA, Axcrona K, Axcrona U, Bigalke I, Brennhovd B, Inderberg EM, Hønnåshagen TK, Skoge LJ, Solum G, Saebøe-Larssen S, Josefsen D, Olaussen RW, Aamdal S, Skotheim RI, Myklebust TÅ, Schendel DJ, Lilleby W, Dueland S, Kvalheim G. Long-term first-in-man Phase I/II study of an adjuvant dendritic cell vaccine in patients with high-risk prostate cancer after radical prostatectomy. Prostate 2022; 82:245-253. [PMID: 34762317 DOI: 10.1002/pros.24267] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 11/02/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Patients with high-risk prostate cancer (PC) can experience biochemical relapse (BCR), despite surgery, and develop noncurative disease. The present study aimed to reduce the risk of BCR with a personalized dendritic cell (DC) vaccine, given as adjuvant therapy, after robot-assisted laparoscopic prostatectomy (RALP). METHODS Twelve weeks after RALP, 20 patients with high-risk PC and undetectable PSA received DC vaccinations for 3 years or until BCR. The primary endpoint was the time to BCR. The immune response was assessed 7 weeks after surgery (baseline) and at one-time point during the vaccination period. RESULTS Among 20 patients, 11 were BCR-free over a median of 96 months (range: 84-99). The median time from the end of vaccinations to the last follow-up was 57 months (range: 45-60). Nine patients developed BCR, either during (n = 4) or after (n = 5) the vaccination period. Among five patients diagnosed with intraductal carcinoma, three experienced early BCR during the vaccination period. All patients that developed BCR remained in stable disease within a median of 99 months (range: 74-99). The baseline immune response was significantly associated with the immune response during the vaccination period (p = 0.015). For patients diagnosed with extraprostatic extension (EPE), time to BCR was longer in vaccine responders than in non-responders (p = 0.09). Among 12 patients with the International Society of Urological Pathology (ISUP) grade 5 PC, five achieved remission after 84 months, and all mounted immune responses. CONCLUSION Patients diagnosed with EPE and ISUP grade 5 PC were at particularly high risk of developing postsurgical BCR. In this subgroup, the vaccine response was related to a reduced BCR incidence. The vaccine was safe, without side effects. This adjuvant first-in-man Phase I/II DC vaccine study showed promising results. DC vaccines after curative surgery should be investigated further in a larger cohort of patients with high-risk PC.
Collapse
Affiliation(s)
| | - Karol Axcrona
- Department of Urology, Oslo University Hospital HF, Oslo, Norway
- Department of Urology, Akershus University Hospital HF, Oslo, Norway
| | - Ulrika Axcrona
- Department of Pathology, Oslo University Hospital HF, Oslo, Norway
| | - Iris Bigalke
- Department of Oncology, Oslo University Hospital HF, Oslo, Norway
- BioNTech IMFS GmbH, Idar-Oberstein, Germany
| | - Bjørn Brennhovd
- Department of Urology, Oslo University Hospital HF, Oslo, Norway
| | - Else M Inderberg
- Department of Oncology, Oslo University Hospital HF, Oslo, Norway
| | | | - Lisbeth J Skoge
- Department of Oncology, Oslo University Hospital HF, Oslo, Norway
| | - Guri Solum
- Department of Oncology, Oslo University Hospital HF, Oslo, Norway
| | | | - Dag Josefsen
- Department of Oncology, Oslo University Hospital HF, Oslo, Norway
| | | | - Steinar Aamdal
- Department for Clinical Research, Oslo University Hospital HF, Oslo, Norway
| | - Rolf I Skotheim
- Department of Molecular Oncology, Oslo University Hospital HF, Oslo, Norway
| | - Tor Å Myklebust
- Department of Registration, Cancer Registry Norway, Oslo, Norway
- Department of Research and Innovation, Møre and Romsdal Hospital Trust, Ålesund, Norway
| | | | - Wolfgang Lilleby
- Department of Oncology, Oslo University Hospital HF, Oslo, Norway
| | - Svein Dueland
- Department for Clinical Research, Oslo University Hospital HF, Oslo, Norway
| | - Gunnar Kvalheim
- Department of Oncology, Oslo University Hospital HF, Oslo, Norway
| |
Collapse
|
14
|
Li Y, Dai X, He L, Bu Y, Ao JP. Crystal-reconstructed BiVO 4 semiconductor photoelectrochemical sensor for ultra-sensitive tumor biomarker detection. J Mater Chem B 2022; 10:870-879. [PMID: 35050300 DOI: 10.1039/d1tb02576g] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In this study, we developed a crystal-reconstructed-BiVO4 aptamer photoelectrochemical (PEC) biosensor by a high-energy laser treatment technique. This biosensor achieves a limit of detection (LOD) (0.82 ag mL-1), linear detection range (1 ag mL-1 to 2 ng mL-1), and resolution ratio (∼18 molecules per mL) for prostate-specific antigen (PSA) tumor biomarker detection. Furthermore, reconstructed surface microstructure and oxygen vacancy doping energy formation after crystal reconstruction induce the stereo-hindrance effect and photogenerated hole energy is reduced during PSA target detection. In this case, a photocurrent inhibition phenomenon for PSA detection is noticed. Based on this photocurrent inversion phenomenon, some dysoxidizable nucleonic acid tumor (miRNA-21) and virus biomarkers (RdRp-COVID) can be detected with a LOD level of ∼10-16 M by linking the corresponding base paring probe on the surface of the crystal-reconstructed photoanode. In addition to high sensitivity, this PEC biosensor presents high detection specificity, stability, and accuracy in clinical verification. Thus, this crystal-reconstructed PEC biosensor shows application potential in the fields of multi-tumor or viral biomarker detection.
Collapse
Affiliation(s)
- Yang Li
- Key Laboratory of Wide Band-Gap Semiconductor Materials and Devices, School of Microelectronics, Xidian University, Xi'an, 710071, China.
| | - Xianying Dai
- Key Laboratory of Wide Band-Gap Semiconductor Materials and Devices, School of Microelectronics, Xidian University, Xi'an, 710071, China.
| | - Lin He
- Shaanxi Provincial Cancer Hospital, Xi'an, 710061, China
| | - Yuyu Bu
- Key Laboratory of Wide Band-Gap Semiconductor Materials and Devices, School of Microelectronics, Xidian University, Xi'an, 710071, China.
| | - Jin-Ping Ao
- Key Laboratory of Wide Band-Gap Semiconductor Materials and Devices, School of Microelectronics, Xidian University, Xi'an, 710071, China.
| |
Collapse
|
15
|
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
|
16
|
Genomic Features and Clinical Implications of Intraductal Carcinoma of the Prostate. Int J Mol Sci 2021; 22:ijms222313125. [PMID: 34884926 PMCID: PMC8658449 DOI: 10.3390/ijms222313125] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/02/2021] [Accepted: 12/02/2021] [Indexed: 01/29/2023] Open
Abstract
Intraductal carcinoma of the prostate (IDC-P) is a rare and unique form of aggressive prostate carcinoma, which is characterized by an expansile proliferation of malignant prostatic epithelial cells within prostatic ducts or acini and the preservation of basal cell layers around the involved glands. The vast majority of IDC-P tumors result from adjacent high-grade invasive cancer via the retrograde spreading of tumor cells into normal prostatic ducts or acini. A subset of IDC-P tumors is rarely derived from the de novo intraductal proliferation of premalignant cells. The presence of IDC-P in biopsy or surgical specimens is significantly associated with aggressive pathologic features, such as high Gleason grade, large tumor volume, and advanced tumor stage, and with poor clinical courses, including earlier biochemical recurrence, distant metastasis, and worse survival outcomes. These architectural and behavioral features of IDC-P may be driven by specific molecular properties. Notably, IDC-P possesses distinct genomic profiles, including higher rates of TMPRSS2–ERG gene fusions and PTEN loss, increased percentage of genomic instability, and higher prevalence of germline BRCA2 mutations. Considering that IDC-P tumors are usually resistant to conventional therapies for prostate cancer, further studies should be performed to develop optimal therapeutic strategies based on distinct genomic features, such as treatment with immune checkpoint blockades or poly (adenosine diphosphate–ribose) polymerase inhibitors for patients harboring increased genomic instability or BRCA2 mutations, as well as genetic counseling with genetic testing. Patient-derived xenografts and tumor organoid models can be the promising in vitro platforms for investigating the molecular features of IDC-P tumor.
Collapse
|
17
|
Considine B, Adeniran A, Hurwitz ME. Current Understanding and Management of Intraductal Carcinoma of the Prostate. Curr Oncol Rep 2021; 23:110. [PMID: 34272624 DOI: 10.1007/s11912-021-01090-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE OF REVIEW This review will discuss current understanding and management approaches of Intraductal carcinoma of the prostate (IDC-P). IDC-P is a histological finding characterized by neoplastic cells that expand but do not invade prostate ducts. RECENT FINDINGS The presence of IDC-P on a prostate biopsy is almost always associated with an invasive disease component and is independently associated with worse clinical outcomes in both early and late disease. These tumors are enriched for mutations in homologous DNA recombination repair (HRR) leading to high genomic instability. Multiparametric MRI with targeted biopsy may aid in diagnosis. Given the poor clinical outcomes associated with this histologic entity, its presence in biopsies should warrant consideration of aggressive management.
Collapse
Affiliation(s)
- Bryden Considine
- Yale Comprehensive Cancer Center, 333 Cedar St, New Haven, CT, 06510, USA
| | - Adebowale Adeniran
- Yale Comprehensive Cancer Center, 333 Cedar St, New Haven, CT, 06510, USA
| | - Michael E Hurwitz
- Yale Comprehensive Cancer Center, 333 Cedar St, New Haven, CT, 06510, USA.
| |
Collapse
|
18
|
Abstract
Intraductal carcinoma of the prostate gland (IDCP) is characterized by an expansile, architecturally, and cytologically atypical proliferation of prostatic epithelial cells within preexisting prostatic ducts and acini. There has been a wider recognition of IDCP by practicing pathologists since its recognition as a separate category in the World Health Organization (WHO) 2016 classification of tumours of the prostate gland. However, there is also a lack of clarity regarding the diagnosis and reporting of IDCP, which has been compounded by divergent expert recommendations regarding the grading of invasive prostate cancers associated with an intraductal component. The International Society of Urological Pathologists (ISUP) recommends that the IDCP component should be incorporated into the Gleason score, while the Genitourinary Pathology Society (GUPS) recommends excluding it when grading prostate cancer. This review seeks to clarify some of these issues and outline a pragmatic approach to reporting IDCP, particularly in needle biopsies. Diagnostic issues and terminology for lesions falling short of IDCP but exceeding that of high-grade prostatic intraepithelial neoplasia are discussed. The management of patients whose prostate biopsies show only IDCP without an associated invasive component is controversial. Some experts recommend radical therapy, while others recommend prompt repeat biopsy. An alternative clinicopathologic approach that takes into consideration the extent, histomorphology, and location (with respect to a radiologic abnormality) of IDCP, as well as radiologic features, is outlined.
Collapse
Affiliation(s)
- Murali Varma
- Department of Cellular Pathology, University Hospital of Wales, Cardiff, UK
| |
Collapse
|
19
|
Choo N, Ramm S, Luu J, Winter JM, Selth LA, Dwyer AR, Frydenberg M, Grummet J, Sandhu S, Hickey TE, Tilley WD, Taylor RA, Risbridger GP, Lawrence MG, Simpson KJ. High-Throughput Imaging Assay for Drug Screening of 3D Prostate Cancer Organoids. SLAS DISCOVERY 2021; 26:1107-1124. [PMID: 34111999 PMCID: PMC8458687 DOI: 10.1177/24725552211020668] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
New treatments are required for advanced prostate cancer; however, there are fewer preclinical models of prostate cancer than other common tumor types to test candidate therapeutics. One opportunity to increase the scope of preclinical studies is to grow tissue from patient-derived xenografts (PDXs) as organoid cultures. Here we report a scalable pipeline for automated seeding, treatment and an analysis of the drug responses of prostate cancer organoids. We established organoid cultures from 5 PDXs with diverse phenotypes of prostate cancer, including castrate-sensitive and castrate-resistant disease, as well as adenocarcinoma and neuroendocrine pathology. We robotically embedded organoids in Matrigel in 384-well plates and monitored growth via brightfield microscopy before treatment with poly ADP-ribose polymerase inhibitors or a compound library. Independent readouts including metabolic activity and live-cell imaging–based features provided robust measures of organoid growth and complementary ways of assessing drug efficacy. Single organoid analyses enabled in-depth assessment of morphological differences between patients and within organoid populations and revealed that larger organoids had more striking changes in morphology and composition after drug treatment. By increasing the scale and scope of organoid experiments, this automated assay complements other patient-derived models and will expedite preclinical testing of new treatments for prostate cancer.
Collapse
Affiliation(s)
- Nicholas Choo
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Susanne Ramm
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.,Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Jennii Luu
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Jean M Winter
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.,Freemason's Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, SA, Australia
| | - Luke A Selth
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.,Freemason's Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, SA, Australia.,Flinders Health and Medical Research Institute, Flinders University, Adelaide, SA, Australia
| | - Amy R Dwyer
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Mark Frydenberg
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Australian Urology Associates, Melbourne, VIC, Australia.,Department of Urology, Cabrini Health, Malvern, VIC, Australia
| | - Jeremy Grummet
- Australian Urology Associates, Melbourne, VIC, Australia.,Epworth Healthcare, Melbourne, VIC, Australia.,Department of Surgery, Central Clinical School, Monash University, Clayton, VIC, Australia
| | - Shahneen Sandhu
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.,Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Cancer Tissue Collection After Death (CASCADE) Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Theresa E Hickey
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Wayne D Tilley
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.,Freemason's Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, SA, Australia
| | - Renea A Taylor
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.,Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Physiology, Monash University, Clayton, VIC, Australia.,Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Melbourne Urological Research Alliance (MURAL), Monash Biomedicine Discovery Institute Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Gail P Risbridger
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.,Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Melbourne Urological Research Alliance (MURAL), Monash Biomedicine Discovery Institute Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Mitchell G Lawrence
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.,Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Melbourne Urological Research Alliance (MURAL), Monash Biomedicine Discovery Institute Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Kaylene J Simpson
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.,Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| |
Collapse
|
20
|
Erickson A, Hayes A, Rajakumar T, Verrill C, Bryant RJ, Hamdy FC, Wedge DC, Woodcock DJ, Mills IG, Lamb AD. A Systematic Review of Prostate Cancer Heterogeneity: Understanding the Clonal Ancestry of Multifocal Disease. Eur Urol Oncol 2021; 4:358-369. [PMID: 33888445 DOI: 10.1016/j.euo.2021.02.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/31/2021] [Accepted: 02/26/2021] [Indexed: 11/24/2022]
Abstract
CONTEXT Studies characterising genomic changes in prostate cancer (PCa) during natural progression have greatly increased our understanding of the disease. A better understanding of the evolutionary history of PCa would allow advances in diagnostics, prognostication, and novel therapies that together will improve patient outcomes. OBJECTIVE To review the molecular heterogeneity of PCa and assess recent efforts to profile intratumoural heterogeneity and clonal evolution. EVIDENCE ACQUISITION We screened a total of 1313 abstracts from PubMed published between 2009 and 2020, of which we reviewed 84 full-text articles. We excluded 49, resulting in 35 studies for qualitative analysis. EVIDENCE SYNTHESIS In studies of primary disease (16 studies, 4793 specimens), there is a lack of consensus regarding the monoclonal or polyclonal origin of primary PCa. There is no consistent mutation giving rise to primary PCa. Detailed clonal analysis of primary PCa has been limited by current techniques. By contrast, clonal relationships between PCa metastases and a potentiating clone have been consistently identified (19 studies, 732 specimens). Metastatic specimens demonstrate consistent truncal genomic aberrations that suggest monoclonal metastatic progenitors. CONCLUSIONS The relationship between the clonal dynamics of PCa and clinical outcomes needs further investigation. It is likely that this will provide a biological rationale for whether radical treatment of the primary tumour benefits patients with oligometastatic PCa. Future studies on the mutational burden in primary disease at single-cell resolution should permit the identification of clonal patterns underpinning the origin of lethal PCa. PATIENT SUMMARY Prostate cancers arise in different parts of the prostate because of DNA mutations that occur by chance at different times. These cancer cells and their origin can be tracked by DNA mapping. In this review we summarise the state of the art and outline what further science is needed to provide the missing answers.
Collapse
Affiliation(s)
- Andrew Erickson
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Alicia Hayes
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK; Oxford NIHR Biomedical Research Centre, University of Oxford, UK
| | - Timothy Rajakumar
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Clare Verrill
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK; Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Oxford National Institute for Health Research Biomedical Research Centre, Oxford, UK; Oxford NIHR Biomedical Research Centre, University of Oxford, UK
| | - Richard J Bryant
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK; Department of Urology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Oxford NIHR Biomedical Research Centre, University of Oxford, UK
| | - Freddie C Hamdy
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK; Department of Urology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Oxford NIHR Biomedical Research Centre, University of Oxford, UK
| | - David C Wedge
- Manchester Cancer Research Centre, University of Manchester, Manchester, UK
| | - Dan J Woodcock
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK; Oxford Big Data Institute, University of Oxford, Oxford, UK
| | - Ian G Mills
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK; Oxford NIHR Biomedical Research Centre, University of Oxford, UK
| | - Alastair D Lamb
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK; Department of Urology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Oxford NIHR Biomedical Research Centre, University of Oxford, UK.
| |
Collapse
|
21
|
Shojo K, Kosaka T, Nakamura K, Hongo H, Kobayashi H, Mikami S, Nishihara H, Oya M. First case of ductal adenocarcinoma of the prostate with MAP3K1 homozygous deletion. IJU Case Rep 2021; 4:176-179. [PMID: 33977253 PMCID: PMC8088887 DOI: 10.1002/iju5.12274] [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: 01/06/2021] [Revised: 02/08/2021] [Accepted: 02/14/2021] [Indexed: 11/29/2022] Open
Abstract
INTRODUCTION Ductal adenocarcinoma of the prostate is a rare prostate cancer variant and associated with higher stage and greater risk of mortality. Optimal systemic therapy for metastatic ductal adenocarcinoma is not known. CASE PRESENTATION A 67-year-old man presented with ductal adenocarcinoma of the prostate accompanied by multiple lung metastases and advanced bone metastases. We performed channel transurethral resection of the prostate and confirmed the diagnosis of ductal adenocarcinoma of the prostate. DNA sequencing identified a TP53 somatic point mutation (p.Gly245Ser) as the pathogenic variant. Furthermore, a homozygous deletion was observed in mitogen-activated protein kinase kinase kinase 1. The patient received enzalutamide but deceased 5 months after presenting to our institution. CONCLUSION To our knowledge, this is the first report of ductal adenocarcinoma of the prostate with a mitogen-activated protein kinase kinase kinase 1 homozygous deletion. Accumulation of whole-exome sequencing data is expected to inform future advances in therapy development.
Collapse
Affiliation(s)
- Kazunori Shojo
- Department of UrologyKeio University School of MedicineTokyoJapan
| | - Takeo Kosaka
- Department of UrologyKeio University School of MedicineTokyoJapan
| | - Kohei Nakamura
- Genomics UnitKeio Cancer CenterKeio University School of MedicineTokyoJapan
| | - Hiroshi Hongo
- Department of UrologyKeio University School of MedicineTokyoJapan
| | | | - Shuji Mikami
- Division of Diagnostic PathologyKeio University HospitalTokyoJapan
| | - Hiroshi Nishihara
- Genomics UnitKeio Cancer CenterKeio University School of MedicineTokyoJapan
| | - Mototsugu Oya
- Department of UrologyKeio University School of MedicineTokyoJapan
| |
Collapse
|
22
|
Chow K, Bedő J, Ryan A, Agarwal D, Bolton D, Chan Y, Dundee P, Frydenberg M, Furrer MA, Goad J, Gyomber D, Hanegbi U, Harewood L, King D, Lamb AD, Lawrentschuk N, Liodakis P, Moon D, Murphy DG, Peters JS, Ruljancich P, Verrill CL, Webb D, Wong LM, Zargar H, Costello AJ, Papenfuss AT, Hovens CM, Corcoran NM. Ductal variant prostate carcinoma is associated with a significantly shorter metastasis-free survival. Eur J Cancer 2021; 148:440-450. [PMID: 33678516 DOI: 10.1016/j.ejca.2020.12.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/09/2020] [Accepted: 12/19/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND Ductal adenocarcinoma is an uncommon prostate cancer variant. Previous studies suggest that ductal variant histology may be associated with worse clinical outcomes, but these are difficult to interpret. To address this, we performed an international, multi-institutional study to describe the characteristics of ductal adenocarcinoma, particularly focussing on the effect of presence of ductal variant cancer on metastasis-free survival. METHODS Patients with ductal variant histology from two institutional databases who underwent radical prostatectomies were identified and compared with an independent acinar adenocarcinoma cohort. After propensity score matching, the effect of the presence of ductal adenocarcinoma on time to biochemical recurrence, initiation of salvage therapy and the development of metastatic disease was determined. Deep whole-exome sequencing was performed for selected cases (n = 8). RESULTS A total of 202 ductal adenocarcinoma and 2037 acinar adenocarcinoma cases were analysed. Survival analysis after matching demonstrated that patients with ductal variant histology had shorter salvage-free survival (8.1 versus 22.0 months, p = 0.03) and metastasis-free survival (6.7 versus 78.6 months, p < 0.0001). Ductal variant histology was consistently associated with RB1 loss, as well as copy number gains in TAP1, SLC4A2 and EHHADH. CONCLUSIONS The presence of any ductal variant adenocarcinoma at the time of prostatectomy portends a worse clinical outcome than pure acinar cancers, with significantly shorter times to initiation of salvage therapies and the onset of metastatic disease. These features appear to be driven by uncoupling of chromosomal duplication from cell division, resulting in widespread copy number aberration with specific gain of genes implicated in treatment resistance.
Collapse
Affiliation(s)
- Ken Chow
- Department of Surgery, University of Melbourne, Parkville, Victoria, Australia; Urology Unit, Royal Melbourne Hospital, Parkville, Victoria, Australia.
| | - Justin Bedő
- Bioinformatics Division, Walter & Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia; Department of Computing and Information Systems, University of Melbourne, Melbourne, Victoria, Australia
| | - Andrew Ryan
- TissuPath Specialist Pathology, Mount Waverley, Victoria, Australia
| | - Dinesh Agarwal
- Urology Unit, Royal Melbourne Hospital, Parkville, Victoria, Australia; Department of Urology, Western Health, Footscray, Victoria, Australia
| | - Damien Bolton
- Department of Urology, Austin Health, Heidelberg, Victoria, Australia
| | - Yee Chan
- Department of Urology, Austin Health, Heidelberg, Victoria, Australia
| | - Philip Dundee
- Department of Surgery, University of Melbourne, Parkville, Victoria, Australia; Urology Unit, Royal Melbourne Hospital, Parkville, Victoria, Australia; Department of Urology, Western Health, Footscray, Victoria, Australia
| | - Mark Frydenberg
- Department of Surgery, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia; Cabrini Institute, Cabrini Health, Malvern, Victoria, Australia
| | - Marc A Furrer
- Urology Unit, Royal Melbourne Hospital, Parkville, Victoria, Australia; Department of Urology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Jeremy Goad
- Genitourinary Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Department of Urology, St Vincent's Health, Fitzroy, Victoria, Australia
| | - Dennis Gyomber
- Department of Urology, Austin Health, Heidelberg, Victoria, Australia
| | - Uri Hanegbi
- Department of Urology, Alfred Health, Melbourne, Victoria, Australia
| | - Laurence Harewood
- Department of Surgery, University of Melbourne, Parkville, Victoria, Australia; Urology Unit, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Dennis King
- Department of Urology, Alfred Health, Melbourne, Victoria, Australia
| | - Alastair D Lamb
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Nathan Lawrentschuk
- Urology Unit, Royal Melbourne Hospital, Parkville, Victoria, Australia; Department of Urology, Austin Health, Heidelberg, Victoria, Australia; Genitourinary Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Peter Liodakis
- Department of Urology, Austin Health, Heidelberg, Victoria, Australia
| | - Daniel Moon
- Department of Surgery, University of Melbourne, Parkville, Victoria, Australia; Urology Unit, Royal Melbourne Hospital, Parkville, Victoria, Australia; Genitourinary Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Declan G Murphy
- Genitourinary Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Justin S Peters
- Urology Unit, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | | | - Clare L Verrill
- Department of Pathology, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - David Webb
- Department of Urology, Austin Health, Heidelberg, Victoria, Australia
| | - Lih-Ming Wong
- Department of Surgery, University of Melbourne, Parkville, Victoria, Australia; Department of Urology, Austin Health, Heidelberg, Victoria, Australia; Department of Urology, St Vincent's Health, Fitzroy, Victoria, Australia
| | - Homayoun Zargar
- Department of Surgery, University of Melbourne, Parkville, Victoria, Australia; Urology Unit, Royal Melbourne Hospital, Parkville, Victoria, Australia; Department of Urology, Western Health, Footscray, Victoria, Australia
| | - Anthony J Costello
- Department of Surgery, University of Melbourne, Parkville, Victoria, Australia; Urology Unit, Royal Melbourne Hospital, Parkville, Victoria, Australia; Australian Prostate Centre, North Melbourne, Victoria, Australia
| | - Anthony T Papenfuss
- Bioinformatics Division, Walter & Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia; Victorian Comprehensive Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia; School of Mathematics and Statistics, University of Melbourne, Parkville, Victoria, Australia
| | - Christopher M Hovens
- Department of Surgery, University of Melbourne, Parkville, Victoria, Australia; Victorian Comprehensive Cancer Centre, Melbourne, Victoria, Australia
| | - Niall M Corcoran
- Department of Surgery, University of Melbourne, Parkville, Victoria, Australia; Urology Unit, Royal Melbourne Hospital, Parkville, Victoria, Australia; Victorian Comprehensive Cancer Centre, Melbourne, Victoria, Australia; Department of Urology, Frankston Hospital, Frankston, Victoria, Australia
| |
Collapse
|
23
|
Ranasinghe W, Shapiro DD, Zhang M, Bathala T, Navone N, Thompson TC, Broom B, Aparicio A, Tu SM, Tang C, Davis JW, Pisters L, Chapin BF. Optimizing the diagnosis and management of ductal prostate cancer. Nat Rev Urol 2021; 18:337-358. [PMID: 33824525 DOI: 10.1038/s41585-021-00447-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2021] [Indexed: 12/13/2022]
Abstract
Ductal adenocarcinoma (DAC) is the most common variant histological subtype of prostate carcinoma and has an aggressive clinical course. DAC is usually characterized and treated as high-risk prostatic acinar adenocarcinoma (PAC). However, DAC has a different biology to that of acinar disease, which often poses a challenge for both diagnosis and management. DAC can be difficult to identify using conventional diagnostic modalities such as serum PSA levels and multiparametric MRI, and the optimal management for localized DAC is unknown owing to the rarity of the disease. Following definitive therapy for localized disease with radical prostatectomy or radiotherapy, the majority of DACs recur with visceral metastases at low PSA levels. Various systemic therapies that have been shown to be effective in high-risk PAC have limited use in treating DAC. Although current understanding of the biology of DAC is limited, genomic analyses have provided insights into the pathology behind its aggressive behaviour and potential future therapeutic targets.
Collapse
Affiliation(s)
- Weranja Ranasinghe
- Department of Urology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
| | - Daniel D Shapiro
- Department of Urology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Miao Zhang
- Department of Pathology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Tharakeswara Bathala
- Department of Radiology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Nora Navone
- Department of Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Timothy C Thompson
- Department of Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Bradley Broom
- Department of Bioinformatics and Computational Biology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Ana Aparicio
- Department of Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Shi-Ming Tu
- Department of Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Chad Tang
- Department of Radiation Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - John W Davis
- Department of Urology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Louis Pisters
- Department of Urology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Brian F Chapin
- Department of Urology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
24
|
Patel HRH, Patel R. Are we progressing in prostate cancer management? Asian J Urol 2021; 8:241-242. [PMID: 33996484 PMCID: PMC8099682 DOI: 10.1016/j.ajur.2020.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 07/21/2020] [Indexed: 12/24/2022] Open
Affiliation(s)
- Hiten RH. Patel
- Department of Urology, University of Rochester Medical & Dental School, NY, USA
| | - Reena Patel
- St George's University of London Medical School, London, United Kingdom
| |
Collapse
|
25
|
Porter LH, Bakshi A, Pook D, Clark A, Clouston D, Kourambas J, Goode DL, Risbridger GP, Taylor RA, Lawrence MG. Androgen receptor enhancer amplification in matched patient-derived xenografts of primary and castrate-resistant prostate cancer. J Pathol 2021; 254:121-134. [PMID: 33620092 DOI: 10.1002/path.5652] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/01/2021] [Accepted: 02/17/2021] [Indexed: 12/30/2022]
Abstract
Amplifications of the androgen receptor (AR) occur in up to 80% of men with castration-resistant prostate cancer (CRPC). Recent studies highlighted that these amplifications not only span the AR gene but usually encompass a distal enhancer. This represents a newly recognised, non-coding mechanism of resistance to AR-directed therapies, including enzalutamide. To study disease progression before and after AR amplification, we used tumour samples from a castrate-sensitive primary tumour and castrate-resistant metastasis of the same patient. For subsequent functional and genomic studies, we established serially transplantable patient-derived xenografts (PDXs). Whole genome sequencing showed that alterations associated with poor prognosis, such as TP53 and PTEN loss, existed before androgen deprivation therapy, followed by co-amplification of the AR gene and enhancer after the development of metastatic CRPC. The PDX of the primary tumour, without the AR amplification, was sensitive to AR-directed treatments, including castration, enzalutamide, and apalutamide. The PDX of the metastasis, with the AR amplification, had higher AR and AR-V7 expression in castrate conditions, and was resistant to castration, apalutamide, and enzalutamide in vivo. Treatment with a BET inhibitor outperformed the AR-directed therapies for the metastasis, resulting in tumour regression for some, but not all, grafts. Therefore, this study provides novel matched PDXs to test potential treatments that target the overabundance of AR in tumours with AR enhancer amplifications. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Laura H Porter
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Andrew Bakshi
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Computational Cancer Biology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - David Pook
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Medical Oncology, Monash Health, Clayton, VIC, Australia
| | - Ashlee Clark
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | | | - John Kourambas
- Department of Medicine, Monash Health, Casey Hospital, Berwick, VIC, Australia
| | -
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Melbourne Urological Research Alliance (MURAL), Biomedicine Discovery Institute Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - David L Goode
- Computational Cancer Biology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Gail P Risbridger
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Renea A Taylor
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.,Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Physiology, Monash University, Clayton, VIC, Australia
| | - Mitchell G Lawrence
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
26
|
Risbridger GP, Lawrence MG, Taylor RA. PDX: Moving Beyond Drug Screening to Versatile Models for Research Discovery. J Endocr Soc 2020; 4:bvaa132. [PMID: 33094211 PMCID: PMC7566391 DOI: 10.1210/jendso/bvaa132] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 09/10/2020] [Indexed: 01/08/2023] Open
Abstract
Patient-derived xenografts (PDXs) are tools of the trade for many researchers from all disciplines and medical specialties. Most endocrinologists, and especially those working in oncology, commonly use PDXs for preclinical drug testing and development, and over the last decade large collections of PDXs have emerged across all tumor streams. In this review, we examine how the field has evolved to include PDXs as versatile resources for research discoveries, providing evidence for guidelines and changes in clinical practice.
Collapse
Affiliation(s)
- Gail P Risbridger
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute Cancer Program, Monash University, Melbourne, Victoria, Australia.,Prostate Cancer Research Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Mitchell G Lawrence
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute Cancer Program, Monash University, Melbourne, Victoria, Australia.,Prostate Cancer Research Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Renea A Taylor
- Prostate Cancer Research Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.,Department of Physiology, Biomedicine Discovery Institute Cancer Program, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|