1
|
Haffner MC, Morris MJ, Ding CKC, Sayar E, Mehra R, Robinson B, True LD, Gleave M, Lotan TL, Aggarwal R, Huang J, Loda M, Nelson PS, Rubin MA, Beltran H. Framework for the Pathology Workup of Metastatic Castration-Resistant Prostate Cancer Biopsies. Clin Cancer Res 2025; 31:466-478. [PMID: 39589343 PMCID: PMC11790385 DOI: 10.1158/1078-0432.ccr-24-2061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/18/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Lineage plasticity and histologic transformation from prostate adenocarcinoma to neuroendocrine (NE) prostate cancer (NEPC) occur in up to 15% to 20% of patients with castration-resistant prostate cancer (CRPC) as a mechanism of treatment resistance and are associated with aggressive disease and poor prognosis. NEPC tumors typically display small cell carcinoma morphology with loss of androgen receptor (AR) expression and gain of NE lineage markers. However, there is a spectrum of phenotypes that are observed during the lineage plasticity process, and the clinical significance of mixed histologies or those that co-express AR and NE markers or lack all markers is not well defined. Translational research studies investigating NEPC have used variable definitions, making clinical trial design challenging. In this manuscript, we discuss the diagnostic workup of metastatic biopsies to help guide the reproducible classification of phenotypic CRPC subtypes. We recommend classifying CRPC tumors based on histomorphology (adenocarcinoma, small cell carcinoma, poorly differentiated carcinoma, other morphologic variant, or mixed morphology) and IHC markers with a priority for AR, NK3 homeobox 1, insulinoma-associated protein 1, synaptophysin, and cell proliferation based on Ki-67 positivity, with additional markers to be considered based on the clinical context. Ultimately, a unified workup of metastatic CRPC biopsies can improve clinical trial design and eventually practice.
Collapse
Affiliation(s)
- Michael C. Haffner
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Michael J. Morris
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chien-Kuang C. Ding
- Department of Anatomic Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Erolcan Sayar
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Rohit Mehra
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, Ann Arbor, MI, USA
- Rogel Cancer Center, Michigan Medicine, Ann Arbor, MI, USA
| | - Brian Robinson
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Lawrence D. True
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Martin Gleave
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Tamara L. Lotan
- Departments of Pathology, Urology, Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Rahul Aggarwal
- Division of Hematology/Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Jiaoti Huang
- Department of Pathology and Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Peter S. Nelson
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Mark A. Rubin
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Bern Center for Precision Medicine, University of Bern and Inselspital, Bern, Switzerland
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
2
|
Petkov VI, Byun JS, Ward KC, Schussler NC, Archer NP, Bentler S, Doherty JA, Durbin EB, Gershman ST, Cheng I, Insaf T, Gonsalves L, Hernandez BY, Koch L, Liu L, Monnereau A, Morawski BM, Schwartz SM, Stroup A, Wiggins C, Wu XC, Bonds S, Negoita S, Penberthy L. Reporting tumor genomic test results to SEER registries via linkages. J Natl Cancer Inst Monogr 2024; 2024:168-179. [PMID: 39102888 DOI: 10.1093/jncimonographs/lgae013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Precision medicine has become a mainstay of cancer care in recent years. The National Cancer Institute (NCI) Surveillance, Epidemiology, and End Results (SEER) Program has been an authoritative source of cancer statistics and data since 1973. However, tumor genomic information has not been adequately captured in the cancer surveillance data, which impedes population-based research on molecular subtypes. To address this, the SEER Program has developed and implemented a centralized process to link SEER registries' tumor cases with genomic test results that are provided by molecular laboratories to the registries. METHODS Data linkages were carried out following operating procedures for centralized linkages established by the SEER Program. The linkages used Match*Pro, a probabilistic linkage software, and were facilitated by the registries' trusted third party (an honest broker). The SEER registries provide to NCI limited datasets that undergo preliminary evaluation prior to their release to the research community. RESULTS Recently conducted genomic linkages included OncotypeDX Breast Recurrence Score, OncotypeDX Breast Ductal Carcinoma in Situ, OncotypeDX Genomic Prostate Score, Decipher Prostate Genomic Classifier, DecisionDX Uveal Melanoma, DecisionDX Preferentially Expressed Antigen in Melanoma, DecisionDX Melanoma, and germline tests results in Georgia and California SEER registries. CONCLUSIONS The linkages of cancer cases from SEER registries with genomic test results obtained from molecular laboratories offer an effective approach for data collection in cancer surveillance. By providing de-identified data to the research community, the NCI's SEER Program enables scientists to investigate numerous research inquiries.
Collapse
Affiliation(s)
- Valentina I Petkov
- Surveillance Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, MD, USA
| | - Jung S Byun
- Surveillance Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, MD, USA
| | | | | | - Natalie P Archer
- Cancer Epidemiology and Surveillance Branch, Texas Department of State Health Services, Austin, TX, USA
| | - Suzanne Bentler
- Iowa Cancer Registry, The University of Iowa, Iowa City, IA, USA
| | - Jennifer A Doherty
- Hunstman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
| | - Eric B Durbin
- Cancer Research Informatics Shared Resource Facility, Markey Cancer Center, Kentucky Cancer Registry, University of Kentucky, Lexington, KY, USA
| | | | - Iona Cheng
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Tabassum Insaf
- New York State Department of Health, New York State Cancer Registry, Albany, NY, USA
| | - Lou Gonsalves
- Connecticut Department of Public Health, Connecticut Tumor Registry, Hartford, CT, USA
| | | | - Lori Koch
- Illinois State Cancer Registry, Springfield, IL, USA
| | - Lihua Liu
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Alain Monnereau
- Public Health Institute, Cancer Registry of Greater California, Sacramento, CA, USA
| | | | - Stephen M Schwartz
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Charles Wiggins
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Xiao-Cheng Wu
- School of Medicine, Louisiana State University, New Orleans, LA, USA
| | - Sarah Bonds
- Surveillance Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, MD, USA
| | - Serban Negoita
- Surveillance Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, MD, USA
| | - Lynne Penberthy
- Surveillance Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
3
|
Ajkunic A, Sayar E, Roudier MP, Patel RA, Coleman IM, De Sarkar N, Hanratty B, Adil M, Zhao J, Zaidi S, True LD, Sperger JM, Cheng HH, Yu EY, Montgomery RB, Hawley JE, Ha G, Persse T, Galipeau P, Lee JK, Harmon SA, Corey E, Lang JM, Sawyers CL, Morrissey C, Schweizer MT, Gulati R, Nelson PS, Haffner MC. Assessment of TROP2, CEACAM5 and DLL3 in metastatic prostate cancer: Expression landscape and molecular correlates. NPJ Precis Oncol 2024; 8:104. [PMID: 38760413 PMCID: PMC11101486 DOI: 10.1038/s41698-024-00599-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 05/03/2024] [Indexed: 05/19/2024] Open
Abstract
Therapeutic approaches targeting proteins on the surface of cancer cells have emerged as an important strategy for precision oncology. To capitalize on the potential impact of drugs targeting surface proteins, detailed knowledge about the expression patterns of the target proteins in tumor tissues is required. In castration-resistant prostate cancer (CRPC), agents targeting prostate-specific membrane antigen (PSMA) have demonstrated clinical activity. However, PSMA expression is lost in a significant number of CRPC tumors. The identification of additional cell surface targets is necessary to develop new therapeutic approaches. Here, we performed a comprehensive analysis of the expression heterogeneity and co-expression patterns of trophoblast cell-surface antigen 2 (TROP2), delta-like ligand 3 (DLL3), and carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) in CRPC samples from a rapid autopsy cohort. We show that DLL3 and CEACAM5 exhibit the highest expression in neuroendocrine prostate cancer (NEPC), while TROP2 is expressed across different CRPC molecular subtypes, except for NEPC. We further demonstrated that AR alterations were associated with higher expression of PSMA and TROP2. Conversely, PSMA and TROP2 expression was lower in RB1-altered tumors. In addition to genomic alterations, we show a tight correlation between epigenetic states, particularly histone H3 lysine 27 methylation (H3K27me3) at the transcriptional start site and gene body of TACSTD2 (encoding TROP2), DLL3, and CEACAM5, and their respective protein expression in CRPC patient-derived xenografts. Collectively, these findings provide insights into patterns and determinants of expression of TROP2, DLL3, and CEACAM5 with implications for the clinical development of cell surface targeting agents in CRPC.
Collapse
Affiliation(s)
- Azra Ajkunic
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Erolcan Sayar
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Radhika A Patel
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Ilsa M Coleman
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Navonil De Sarkar
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Brian Hanratty
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Mohamed Adil
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jimmy Zhao
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Samir Zaidi
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lawrence D True
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | | | - Heather H Cheng
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Evan Y Yu
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Robert B Montgomery
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jessica E Hawley
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Gavin Ha
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Thomas Persse
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Patricia Galipeau
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - John K Lee
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Stephanie A Harmon
- Artificial Intelligence Resource, Molecular Imaging Branch, NCI, NIH, Bethesda, MD, USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA, USA
| | | | - Charles L Sawyers
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Howard Hughes Medical Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Michael T Schweizer
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Roman Gulati
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Peter S Nelson
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Urology, University of Washington, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Michael C Haffner
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| |
Collapse
|
4
|
Iacobas DA. Advanced Molecular Solutions for Cancer Therapy-The Good, the Bad, and the Ugly of the Biomarker Paradigm. Curr Issues Mol Biol 2024; 46:1694-1699. [PMID: 38534725 DOI: 10.3390/cimb46030109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 02/17/2024] [Indexed: 03/28/2024] Open
Abstract
Identifying the most effective actionable molecules whose "smart" manipulation might selectively kill/slow down/stop the proliferation of cancer cells, with few side effects on the normal cells of the tissue, was for decades the single major objective of countless investigators [...].
Collapse
Affiliation(s)
- Dumitru Andrei Iacobas
- Laboratory of Personalized Genomics, Undergraduate Medical Academy, Prairie View A&M University, Prairie View, TX 77446, USA
| |
Collapse
|
5
|
Timofte AD, Caruntu ID, Covic AC, Hancianu M, Girlescu N, Chifu MB, Giusca SE. Renal Function Parameters in Distinctive Molecular Subtypes of Prostate Cancer. Cancers (Basel) 2023; 15:5013. [PMID: 37894380 PMCID: PMC10605320 DOI: 10.3390/cancers15205013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/01/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Prostate cancer is a prevalent malignancy in male patients, having diverse clinical outcomes. The follow-up of patients diagnosed with prostate cancer involves the evaluation of renal function, because its impairment reduces patient survival rates and adds complexity to their treatment and clinical care. This study aimed to investigate the relationship between renal function parameters and distinctive molecular subtypes of prostate adenocarcinomas, defined by the immunoexpression of the SPINK1, ERG, HOXB13, and TFF3 markers. The study group comprised 72 patients with prostate cancer and associated chronic kidney disease (CKD) who underwent radical prostatectomy. Histopathological, molecular, and renal parameters were analyzed. Patients were categorized based on ERG/SPINK1 and HOXB13/TFF3 status, and correlations with renal function and prognostic grade groups were assessed. The ERG+/SPINK1+ subgroup exhibited significantly higher postoperative CKD stages and serum creatinine levels compared to the ERG+/SPINK1- subgroup. This suggests an intricate relationship between SPINK1 overexpression and renal function dynamics. The HOXB13-/TFF3+ subgroup displayed higher preoperative serum creatinine levels and CKD stages than the HOXB13-/TFF3- subgroup, aligning with TFF3's potential role in renal function. Furthermore, the study revealed associations between CKD stages and prognostic grade groups in different molecular subtypes, pointing out an intricate interplay between renal function and tumor behavior. Although the molecular classification of prostate acinar ADK is not yet implemented, this research underscores the variability of renal function parameters in different molecular subtypes, offering potential insights into patient prognosis.
Collapse
Affiliation(s)
- Andrei Daniel Timofte
- Department of Morpho-Functional Sciences I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (N.G.); (M.B.C.); (S.E.G.)
| | - Irina-Draga Caruntu
- Department of Morpho-Functional Sciences I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (N.G.); (M.B.C.); (S.E.G.)
- Department of Pathology, “Dr. C. I. Parhon” University Hospital, 700503 Iasi, Romania
- Romanian Medical Science Academy, 030171 Bucharest, Romania;
| | - Adrian C. Covic
- Romanian Medical Science Academy, 030171 Bucharest, Romania;
- Romanian Academy of Scientists, 50044 Bucharest, Romania
- Department Medical II, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Department of Nephrology, Dialysis and Renal Transplant Center, “Dr. C. I. Parhon” University Hospital, 700503 Iasi, Romania
| | - Monica Hancianu
- Department of Pharmaceutical Sciences II, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Nona Girlescu
- Department of Morpho-Functional Sciences I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (N.G.); (M.B.C.); (S.E.G.)
| | - Mariana Bianca Chifu
- Department of Morpho-Functional Sciences I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (N.G.); (M.B.C.); (S.E.G.)
| | - Simona Eliza Giusca
- Department of Morpho-Functional Sciences I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (N.G.); (M.B.C.); (S.E.G.)
- Department of Pathology, “Dr. C. I. Parhon” University Hospital, 700503 Iasi, Romania
| |
Collapse
|
6
|
Nakamura Y, Tsuji K, Shiraishi T, Sako S, Ogura R, Taga H, Inoue Y, Ohashi M, Ueda S, Yamada T, Ueda T, Fujihara A, Hongo F, Ukimura O. Novel device for dividing core needle biopsy specimens to provide paired mirror image-like tissues for genetic and pathological tests. Sci Rep 2023; 13:6610. [PMID: 37095303 PMCID: PMC10126131 DOI: 10.1038/s41598-023-33776-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 04/18/2023] [Indexed: 04/26/2023] Open
Abstract
In a world that seeks precision medicine, genetic testing is gaining importance in clinical decision making. We previously reported the utility of a novel tool for longitudinally dividing core needle biopsy (CNB) tissues into two filamentous tissues that can provide paired mirror image-like tissues (mirror-tissues) that spatially match each other. In this study, we investigated its application in gene panel testing in patients who underwent prostate CNB. Four hundred and forty-three biopsy cores were obtained from 40 patients. Of them, 361 biopsy cores (81.5%) were judged by a physician to be appropriate for dividing into two pieces using the new device, of which a histopathological diagnosis was successfully reached in 358 biopsy cores (99.2%). Among them, the quality and quantity of nucleic acid in 16 appropriately divided cores were assessed and found to be sufficient for gene panel testing, and histopathological diagnosis was successfully obtained from the remaining divided cores. The novel device for longitudinally-dividing CNB tissue provided mirror image-like paired-tissues for gene panel and pathology testing. The device might be a promising tool for obtaining genetic and molecular biological information, in addition to histopathological diagnosis, helping to advance personalized medicine.
Collapse
Affiliation(s)
- Yuichi Nakamura
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Keisuke Tsuji
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Takumi Shiraishi
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan.
| | - Satoshi Sako
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Ryota Ogura
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Hideto Taga
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Yuta Inoue
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Munehiro Ohashi
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Saya Ueda
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Takeshi Yamada
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Takashi Ueda
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Atsuko Fujihara
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Fumiya Hongo
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Osamu Ukimura
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| |
Collapse
|
7
|
Alfahed A, Ebili HO, Almoammar NE, Alasiri G, AlKhamees OA, Aldali JA, Al Othaim A, Hakami ZH, Abdulwahed AM, Waggiallah HA. Prognostic Values of Gene Copy Number Alterations in Prostate Cancer. Genes (Basel) 2023; 14:genes14050956. [PMID: 37239316 DOI: 10.3390/genes14050956] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/14/2023] [Accepted: 04/20/2023] [Indexed: 05/28/2023] Open
Abstract
Whilst risk prediction for individual prostate cancer (PCa) cases is of a high priority, the current risk stratification indices for PCa management have severe limitations. This study aimed to identify gene copy number alterations (CNAs) with prognostic values and to determine if any combination of gene CNAs could have risk stratification potentials. Clinical and genomic data of 500 PCa cases from the Cancer Genome Atlas stable were retrieved from the Genomic Data Commons and cBioPortal databases. The CNA statuses of a total of 52 genetic markers, including 21 novel markers and 31 previously identified potential prognostic markers, were tested for prognostic significance. The CNA statuses of a total of 51/52 genetic markers were significantly associated with advanced disease at an odds ratio threshold of ≥1.5 or ≤0.667. Moreover, a Kaplan-Meier test identified 27/52 marker CNAs which correlated with disease progression. A Cox Regression analysis showed that the amplification of MIR602 and deletions of MIR602, ZNF267, MROH1, PARP8, and HCN1 correlated with a progression-free survival independent of the disease stage and Gleason prognostic group grade. Furthermore, a binary logistic regression analysis identified twenty-two panels of markers with risk stratification potentials. The best model of 7/52 genetic CNAs, which included the SPOP alteration, SPP1 alteration, CCND1 amplification, PTEN deletion, CDKN1B deletion, PARP8 deletion, and NKX3.1 deletion, stratified the PCa cases into a localised and advanced disease with an accuracy of 70.0%, sensitivity of 85.4%, specificity of 44.9%, positive predictive value of 71.67%, and negative predictive value of 65.35%. This study validated prognostic gene level CNAs identified in previous studies, as well as identified new genetic markers with CNAs that could potentially impact risk stratification in PCa.
Collapse
Affiliation(s)
- Abdulaziz Alfahed
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Henry Okuchukwu Ebili
- Department of Morbid Anatomy and Histopathology, Olabisi Onabanjo University, Ago-Iwoye P.M.B. 2002, Nigeria
| | - Nasser Eissa Almoammar
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Glowi Alasiri
- Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud University, Riyadh 13317, Saudi Arabia
| | - Osama A AlKhamees
- Department of Pharmacology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13317, Saudi Arabia
| | - Jehad A Aldali
- Department of Pathology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13317, Saudi Arabia
| | - Ayoub Al Othaim
- Department of Medical Laboratories, College of Applied Medical Sciences, Majmaah University, Al-Majmaah 11952, Saudi Arabia
| | - Zaki H Hakami
- Medical Laboratory Technology Department, College of Applied Medical Sciences, Jazan University, Jazan 82817, Saudi Arabia
| | - Abdulhadi M Abdulwahed
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11362, Saudi Arabia
| | - Hisham Ali Waggiallah
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| |
Collapse
|
8
|
Zhang M, Liu Y, Yao J, Wang K, Tu J, Hu Z, Jin Y, Du Y, Sun X, Chen L, Wang Z. Value of machine learning-based transrectal multimodal ultrasound combined with PSA-related indicators in the diagnosis of clinically significant prostate cancer. Front Endocrinol (Lausanne) 2023; 14:1137322. [PMID: 36967794 PMCID: PMC10031096 DOI: 10.3389/fendo.2023.1137322] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 02/20/2023] [Indexed: 03/11/2023] Open
Abstract
Objective To investigate the effect of transrectal multimodal ultrasound combined with serum prostate-specific antigen (PSA)-related indicators and machine learning for the diagnosis of clinically significant prostate cancer. Methods Based on Gleason score of postoperative pathological results, the subjects were divided into clinically significant prostate cancer groups(GS>6)and non-clinically significant prostate cancer groups(GS ≤ 6). The independent risk factors were obtained by univariate logistic analysis. Artificial neural network (ANN), logistic regression (LR), support vector machine (SVM), decision tree (DT), random forest (RF), and K-nearest neighbor (KNN) machine learning models were combined with clinically significant prostate cancer risk factors to establish the machine learning model, calculate the model evaluation indicators, construct the receiver operating characteristic curve (ROC), and calculate the area under the curve (AUC). Results Independent risk factor items (P< 0.05) were entered into the machine learning model. A comparison of the evaluation indicators of the model and the area under the ROC curve showed the ANN model to be best at predicting clinically significant prostate cancer, with a sensitivity of 80%, specificity of 88.6%, F1 score of 0.897, and the AUC was 0.855. Conclusion Establishing a machine learning model by rectal multimodal ultrasound and combining it with PSA-related indicators has definite application value in predicting clinically significant prostate cancer.
Collapse
Affiliation(s)
- Maoliang Zhang
- Department of Ultrasound, The Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, China
| | - Yuanzhen Liu
- Department of Ultrasound, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jincao Yao
- Department of Ultrasound, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Kai Wang
- Department of Ultrasound, The Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, China
| | - Jing Tu
- Department of Ultrasound, The Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, China
| | - Zhengbiao Hu
- Department of Ultrasound, The Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, China
| | - Yun Jin
- Department of Ultrasound, The Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, China
| | - Yue Du
- Department of Ultrasound, The Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, China
| | - Xingbo Sun
- Department of Ultrasound, The Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, China
| | - Liyu Chen
- Department of Ultrasound, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Zhengping Wang
- Department of Ultrasound, The Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, China
| |
Collapse
|
9
|
Bakbak H, Sayar E, Kaur HB, Salles DC, Patel RA, Hicks J, Lotan TL, De Marzo AM, Gulati R, Epstein JI, Haffner MC. Clonal relationships of adjacent Gleason pattern 3 and Gleason pattern 5 lesions in Gleason Scores 3+5=8 and 5+3=8. Hum Pathol 2022; 130:18-24. [PMID: 36309296 PMCID: PMC10542864 DOI: 10.1016/j.humpath.2022.10.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/13/2022] [Accepted: 10/20/2022] [Indexed: 11/04/2022]
Abstract
Genomic studies have demonstrated a high level of intra-tumoral heterogeneity in prostate cancer. There is strong evidence suggesting that individual tumor foci can arise as genetically distinct, clonally independent lesions. However, recent studies have also demonstrated that adjacent Gleason pattern (GP) 3 and GP4 lesions can originate from the same clone but follow divergent genetic and morphologic evolution. The clonal relationship of adjacent GP3 and GP5 lesions has thus far not been investigated. Here we analyzed a cohort of 14 cases-11 biopsy and 3 radical prostatectomy specimens-with a Gleason score of 3 + 5 = 8 or 5 + 3 = 8 present in the same biopsy or in a single dominant tumor nodule at radical prostatectomy. Clonal and subclonal relationships between GP3 and GP5 lesions were assessed using genetically validated immunohistochemical assays for ERG, PTEN, and P53. 9/14 (64%) cases showed ERG reactivity in both GP3 and GP5 lesions. Only 1/14 (7%) cases showed a discordant pattern with ERG staining present only in GP3. PTEN expression was lost in 2/14 (14%) cases with perfect concordance between GP5 and GP3. P53 nuclear reactivity was present in 1/14 (7%) case in both GP5 and GP3. This study provides first evidence that the majority of adjacent GP3 and GP5 lesions share driver alterations and are clonally related. In addition, we observed a lower-than-expected rate of PTEN loss in GP5 in the context of Gleason score 3 + 5 = 8 or 5 + 3 = 8 tumors.
Collapse
Affiliation(s)
- Hasim Bakbak
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, 98109, WA, USA
| | - Erolcan Sayar
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, 98109, WA, USA
| | - Harsimar B Kaur
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA
| | - Daniela C Salles
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA
| | - Radhika A Patel
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, 98109, WA, USA
| | - Jessica Hicks
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA
| | - Tamara L Lotan
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA; Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA
| | - Angelo M De Marzo
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA; Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA
| | - Roman Gulati
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, 98109, WA, USA
| | - Jonathan I Epstein
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA; Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA.
| | - Michael C Haffner
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, 98109, WA, USA; Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA; Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, 98109, WA, USA; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, 98195, WA, USA.
| |
Collapse
|
10
|
The Immunotherapy and Immunosuppressive Signaling in Therapy-Resistant Prostate Cancer. Biomedicines 2022; 10:biomedicines10081778. [PMID: 35892678 PMCID: PMC9394279 DOI: 10.3390/biomedicines10081778] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/12/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022] Open
Abstract
Prostate cancer is one of the most common malignant tumors in men. Initially, it is androgen-dependent, but it eventually develops into castration-resistant prostate cancer (CRPC), which is incurable with current androgen receptor signaling target therapy and chemotherapy. Immunotherapy, specifically with immune checkpoint inhibitors, has brought hope for the treatment of this type of prostate cancer. Approaches such as vaccines, adoptive chimeric antigen receptor-T (CAR-T) cells, and immune checkpoint inhibitors have been employed to activate innate and adaptive immune responses to treat prostate cancer, but with limited success. Only Sipuleucel-T and the immune checkpoint inhibitor pembrolizumab are approved by the US FDA for the treatment of limited prostate cancer patients. Prostate cancer has a complex tumor microenvironment (TME) in which various immunosuppressive molecules and mechanisms coexist and interact. Additionally, prostate cancer is considered a “cold” tumor with low levels of tumor mutational burden, low amounts of antigen-presenting and cytotoxic T-cell activation, and high levels of immunosuppressive molecules including cytokines/chemokines. Thus, understanding the mechanisms of immunosuppressive signaling activation and immune evasion will help develop more effective treatments for prostate cancer. The purpose of this review is to summarize emerging advances in prostate cancer immunotherapy, with a particular focus on the molecular mechanisms that lead to immune evasion in prostate cancer. At the same time, we also highlight some potential therapeutic targets to provide a theoretical basis for the treatment of prostate cancer.
Collapse
|
11
|
Kidd SG, Bogaard M, Carm KT, Bakken AC, Maltau AMV, Løvf M, Lothe RA, Axcrona K, Axcrona U, Skotheim RI. In situ
expression of
ERG
protein in the context of tumor heterogeneity identifies prostate cancer patients with inferior prognosis. Mol Oncol 2022; 16:2810-2822. [PMID: 35574900 PMCID: PMC9348599 DOI: 10.1002/1878-0261.13225] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/29/2022] [Accepted: 05/13/2022] [Indexed: 11/12/2022] Open
Abstract
Prognostic biomarkers for prostate cancer are needed to improve prediction of disease course and guide treatment decisions. However, biomarker development is complicated by the common multifocality and heterogeneity of the disease. We aimed to determine the prognostic value of candidate biomarkers transcriptional regulator ERG and related ETS family genes, while considering tumor heterogeneity. In a multisampled, prospective, and treatment‐naïve radical prostatectomy cohort from one tertiary center (2010–2012, median follow‐up 8.1 years), we analyzed ERG protein (480 patients; 2047 tissue cores), and RNA of several ETS genes in a subcohort (165 patients; 778 fresh‐frozen tissue samples). Intra‐ and interfocal heterogeneity was identified in 29% and 33% (ERG protein) and 39% and 27% (ETS RNA) of patients, respectively. ERG protein and ETS RNA was identified exclusively in a nonindex tumor in 31% and 32% of patients, respectively. ERG protein demonstrated independent prognostic value in predicting biochemical (P = 0.04) and clinical recurrence (P = 0.004) and appeared to have greatest prognostic value for patients with Grade Groups 4–5. In conclusion, when heterogeneity is considered, ERG protein is a robust prognostic biomarker for prostate cancer.
Collapse
Affiliation(s)
- Susanne G. Kidd
- Department of Molecular Oncology, Institute for Cancer Research Oslo University Hospital–Radiumhospitalet Oslo Norway
- Institute for Clinical Medicine, Faculty of Medicine University of Oslo Oslo Norway
| | - Mari Bogaard
- Department of Molecular Oncology, Institute for Cancer Research Oslo University Hospital–Radiumhospitalet Oslo Norway
- Institute for Clinical Medicine, Faculty of Medicine University of Oslo Oslo Norway
- Department of Pathology Oslo University Hospital–Radiumhospitalet Oslo Norway
| | - Kristina T. Carm
- Department of Molecular Oncology, Institute for Cancer Research Oslo University Hospital–Radiumhospitalet Oslo Norway
| | - Anne Cathrine Bakken
- Department of Molecular Oncology, Institute for Cancer Research Oslo University Hospital–Radiumhospitalet Oslo Norway
| | - Aase M. V. Maltau
- Department of Molecular Oncology, Institute for Cancer Research Oslo University Hospital–Radiumhospitalet Oslo Norway
| | - Marthe Løvf
- Department of Molecular Oncology, Institute for Cancer Research Oslo University Hospital–Radiumhospitalet Oslo Norway
| | - Ragnhild A. Lothe
- Department of Molecular Oncology, Institute for Cancer Research Oslo University Hospital–Radiumhospitalet Oslo Norway
- Institute for Clinical Medicine, Faculty of Medicine University of Oslo Oslo Norway
| | - Karol Axcrona
- Department of Molecular Oncology, Institute for Cancer Research Oslo University Hospital–Radiumhospitalet Oslo Norway
- Department of Urology Akershus University Hospital Lørenskog Norway
| | - Ulrika Axcrona
- Department of Molecular Oncology, Institute for Cancer Research Oslo University Hospital–Radiumhospitalet Oslo Norway
- Department of Pathology Oslo University Hospital–Radiumhospitalet Oslo Norway
| | - Rolf I. Skotheim
- Department of Molecular Oncology, Institute for Cancer Research Oslo University Hospital–Radiumhospitalet Oslo Norway
- Department of Informatics, Faculty of Mathematics and Natural Sciences University of Oslo Oslo Norway
| |
Collapse
|
12
|
Personalized 3-Gene Panel for Prostate Cancer Target Therapy. Curr Issues Mol Biol 2022; 44:360-382. [PMID: 35723406 PMCID: PMC8929157 DOI: 10.3390/cimb44010027] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/05/2022] [Accepted: 01/11/2022] [Indexed: 11/17/2022] Open
Abstract
Many years and billions spent for research did not yet produce an effective answer to prostate cancer (PCa). Not only each human, but even each cancer nodule in the same tumor, has unique transcriptome topology. The differences go beyond the expression level to the expression control and networking of individual genes. The unrepeatable heterogeneous transcriptomic organization among men makes the quest for universal biomarkers and “fit-for-all” treatments unrealistic. We present a bioinformatics procedure to identify each patient’s unique triplet of PCa Gene Master Regulators (GMRs) and predict consequences of their experimental manipulation. The procedure is based on the Genomic Fabric Paradigm (GFP), which characterizes each individual gene by the independent expression level, expression variability and expression coordination with each other gene. GFP can identify the GMRs whose controlled alteration would selectively kill the cancer cells with little consequence on the normal tissue. The method was applied to microarray data on surgically removed prostates from two men with metastatic PCas (each with three distinct cancer nodules), and DU145 and LNCaP PCa cell lines. The applications verified that each PCa case is unique and predicted the consequences of the GMRs’ manipulation. The predictions are theoretical and need further experimental validation.
Collapse
|