1
|
Mohamed AO, Otifi H, Hassan H, Yousif AA, Mustafa SA, Elsiddig SA, Babker AM, Ali EI, Elhag OO. Exploring the efficacy of AMACR, ERG, and AR immunostains in prostatic adenocarcinoma and their association with novel grade groups. Eur J Histochem 2025; 69:4172. [PMID: 39931952 PMCID: PMC11864098 DOI: 10.4081/ejh.2025.4172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 02/04/2025] [Indexed: 02/28/2025] Open
Abstract
The study examines the utility of AMACR, ERG, and AR immunostains in diagnosing prostatic adenocarcinoma (PCa) and assessing prognosis in comparison to the Gleason score and new WHO grading groups. Seventeen PCa biopsies and five benign prostatic hyperplasia (BPH) biopsies were analyzed. Immunoreactivity, scored from 1 to 3 based on percentage of positive cells and intensity of expression, was assessed, revealing 76.47% positivity for AMACR, 35.29% for ERG, and 94.12% for AR in PCa cases, with variable scores and intensity among markers and grade groups. AMACR sensitivity and ERG specificity were noted. Higher-grade PCa exhibited increased positivity for both markers, indicating prognostic significance. In BPH cases, AMACR showed positivity in 2 cases, ERG in 1, and AR in all cases, albeit with lower expression. Differential expression was observed among immunomarkers and grade groups of malignancy. AMACR and ERG stains serve as sensitive and specific markers for PCa diagnosis and prognosis. Their increasing positivity with higher-grade groups underscores prognostic value. These findings highlight the importance of immunostains in refining PCa diagnosis and prognostication.
Collapse
Affiliation(s)
- Andarawi O. Mohamed
- Pathology Department, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Hassan Otifi
- Pathology Department, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Hesham Hassan
- Pathology Department, College of Medicine, King Khalid University, Abha, Saudi Arabia
- Department of Pathology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Adil A. Yousif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Saadalnour A. Mustafa
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Saudi Arabia
| | - Shawgi A. Elsiddig
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Asaad Ma Babker
- Department of Medical Laboratory Sciences, College of Health Sciences, Gulf Medical University, Ajman, United Arab Emirates
| | - Elryah I. Ali
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Northern Border University, Arar, Saudi Arabia
| | - Omer Osman Elhag
- Department of Histopathology and Cytology, Faculty of Medical Laboratory Sciences, Omdurman Islamic University, Sudan
| |
Collapse
|
2
|
Rajendran R, Beck RC, Waskasi MM, Kelly BD, Bauer DR. Digital analysis of the prostate tumor microenvironment with high-order chromogenic multiplexing. J Pathol Inform 2024; 15:100352. [PMID: 38186745 PMCID: PMC10770522 DOI: 10.1016/j.jpi.2023.100352] [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: 07/18/2023] [Revised: 09/30/2023] [Accepted: 11/16/2023] [Indexed: 01/09/2024] Open
Abstract
As our understanding of the tumor microenvironment grows, the pathology field is increasingly utilizing multianalyte diagnostic assays to understand important characteristics of tumor growth. In clinical settings, brightfield chromogenic assays represent the gold-standard and have developed significant trust as the first-line diagnostic method. However, conventional brightfield tests have been limited to low-order assays that are visually interrogated. We have developed a hybrid method of brightfield chromogenic multiplexing that overcomes these limitations and enables high-order multiplex assays. However, how compatible high-order brightfield multiplexed images are with advanced analytical algorithms has not been extensively evaluated. In the present study, we address this gap by developing a novel 6-marker prostate cancer assay that targets diverse aspects of the tumor microenvironment such as prostate-specific biomarkers (PSMA and p504s), immune biomarkers (CD8 and PD-L1), a prognostic biomarker (Ki-67), as well as an adjunctive diagnostic biomarker (basal cell cocktail) and apply the assay to 143 differentially graded adenocarcinoma prostate tissues. The tissues were then imaged on our spectroscopic multiplexing imaging platform and mined for proteomic and spatial features that were correlated with cancer presence and disease grade. Extracted features were used to train a UMAP model that differentiated healthy from cancerous tissue with an accuracy of 89% and identified clusters of cells based on cancer grade. For spatial analysis, cell-to-cell distances were calculated for all biomarkers and differences between healthy and adenocarcinoma tissues were studied. We report that p504s positive cells were at least 2× closer to cells expressing PD-L1, CD8, Ki-67, and basal cell in adenocarcinoma tissues relative to the healthy control tissues. These findings offer a powerful insight to understand the fingerprint of the prostate tumor microenvironment and indicate that high-order chromogenic multiplexing is compatible with digital analysis. Thus, the presented chromogenic multiplexing system combines the clinical applicability of brightfield assays with the emerging diagnostic power of high-order multiplexing in a digital pathology friendly format that is well-suited for translational studies to better understand mechanisms of tumor development and growth.
Collapse
Affiliation(s)
- Rahul Rajendran
- Roche Diagnostics Solutions, (Ventana Medical Systems, Inc.), Tucson, AZ, USA
| | - Rachel C. Beck
- Roche Diagnostics Solutions, (Ventana Medical Systems, Inc.), Tucson, AZ, USA
| | - Morteza M. Waskasi
- Roche Diagnostics Solutions, (Ventana Medical Systems, Inc.), Tucson, AZ, USA
| | - Brian D. Kelly
- Roche Diagnostics Solutions, (Ventana Medical Systems, Inc.), Tucson, AZ, USA
| | - Daniel R. Bauer
- Roche Diagnostics Solutions, (Ventana Medical Systems, Inc.), Tucson, AZ, USA
| |
Collapse
|
3
|
Liu G, Lin W, Zhang K, Chen K, Niu G, Zhu Y, Liu Y, Li P, Li Z, An Y. Elucidating the prognostic and therapeutic significance of TOP2A in various malignancies. Cancer Genet 2024; 288-289:68-81. [PMID: 39454521 DOI: 10.1016/j.cancergen.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/26/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024]
Abstract
Topoisomerase IIα (TOP2A) is a crucial enzyme that plays a vital role in DNA replication and transcription mechanisms. Dysregulated expression of TOP2A has been associated with various malignancies, including hepatocellular carcinoma, prostate cancer, colon cancer, lung cancer and breast cancer. In this review, we summarized the prognostic relevances of TOP2A in various types of cancer. The increased expression of TOP2A has been linked to resistance to therapy and reduced survival rates. Therefore, evaluating TOP2A levels could assist in identifying patients who may derive advantages from molecular targeted therapy. The amplification of TOP2A has been linked to a positive response to chemotherapy regimens that contain anthracycline. Nevertheless, the overexpression of TOP2A also indicates a heightened likelihood of disease recurrence and unfavorable prognosis. The prognostic significance of TOP2A has been extensively studied in various types of cancer. The increased expression of TOP2A is associated with poor clinical outcomes, indicating its potential as a valuable biomarker for assessing risk and stratifying treatment in these malignancies. However, further investigation is needed to elucidate the underlying mechanisms by which TOP2A influences cancer progression and to explore its potential as a therapeutic target.
Collapse
Affiliation(s)
- Guangchao Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Wenlong Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Kaifeng Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Kangxu Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Guanglin Niu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Yonghao Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Yixuan Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key laboratory of cell signal transduction, Henan University, Kaifeng, 475004, China
| | - Pengkun Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key laboratory of cell signal transduction, Henan University, Kaifeng, 475004, China
| | - Zhihao Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key laboratory of cell signal transduction, Henan University, Kaifeng, 475004, China
| | - Yang An
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key laboratory of cell signal transduction, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
4
|
Hachem S, Yehya A, El Masri J, Mavingire N, Johnson JR, Dwead AM, Kattour N, Bouchi Y, Kobeissy F, Rais-Bahrami S, Mechref Y, Abou-Kheir W, Woods-Burnham L. Contemporary Update on Clinical and Experimental Prostate Cancer Biomarkers: A Multi-Omics-Focused Approach to Detection and Risk Stratification. BIOLOGY 2024; 13:762. [PMID: 39452071 PMCID: PMC11504278 DOI: 10.3390/biology13100762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/11/2024] [Accepted: 09/20/2024] [Indexed: 10/26/2024]
Abstract
Prostate cancer remains a significant health challenge, being the most prevalent non-cutaneous cancer in men worldwide. This review discusses the critical advancements in biomarker discovery using single-omics and multi-omics approaches. Multi-omics, integrating genomic, transcriptomic, proteomic, metabolomic, and epigenomic data, offers a comprehensive understanding of the molecular heterogeneity of prostate cancer, leading to the identification of novel biomarkers and therapeutic targets. This holistic approach not only enhances the specificity and sensitivity of prostate cancer detection but also supports the development of personalized treatment strategies. Key studies highlighted include the identification of novel genes, genetic mutations, peptides, metabolites, and potential biomarkers through multi-omics analyses, which have shown promise in improving prostate cancer management. The integration of multi-omics in clinical practice can potentially revolutionize prostate cancer prognosis and treatment, paving the way for precision medicine. This review underscores the importance of continued research and the application of multi-omics to overcome current challenges in prostate cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Sana Hachem
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon (A.Y.)
| | - Amani Yehya
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon (A.Y.)
| | - Jad El Masri
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon (A.Y.)
| | - Nicole Mavingire
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA 30310, USA; (N.M.)
| | - Jabril R. Johnson
- Department of Microbiology, Biochemistry, & Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA;
| | - Abdulrahman M. Dwead
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA 30310, USA; (N.M.)
| | - Naim Kattour
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon (A.Y.)
| | - Yazan Bouchi
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Firas Kobeissy
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Soroush Rais-Bahrami
- Department of Urology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Radiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- O’Neal Comprehensive Cancer Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409, USA
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon (A.Y.)
| | - Leanne Woods-Burnham
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA 30310, USA; (N.M.)
| |
Collapse
|
5
|
Argalácsová S, Vočka M, Čapoun O, Lambert L. Timing of Early Salvage Therapy for Patients With Biochemical Relapse of Prostate Carcinoma. Oncol Rev 2023; 17:10676. [PMID: 37771544 PMCID: PMC10522833 DOI: 10.3389/or.2023.10676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/30/2023] [Indexed: 09/30/2023] Open
Abstract
Between 25% and 33% of patients after radical prostatectomy experience a relapse of the disease. The risk of relapse increases in patients with risk factors up to 50%-80%. For a long time, adjuvant radiotherapy has been considered the standard of care. Four large prospective trials, that compared adjuvant and salvage radiotherapy in patients with biochemical relapse, showed the superiority of the adjuvant approach in biochemical and local relapse-free survival, but no consistent benefit in long-term endpoints (i.e., metastasis-free survival, overall survival, or carcinoma-specific survival) at the expense of increased urinary and bowel toxicity. Three large international studies comparing adjuvant and salvage radiotherapy paved the way toward early salvage radiotherapy. However, the optimal threshold of the PSA level (range of 0.2-0.5 ng/mL) for initiating early salvage radiotherapy remains unresolved and still poses a challenge in everyday clinical practice when balancing the need for early radiotherapy and the associated toxicity. Imprecise stratification of biochemical relaps patients according to the risk of clinical relapse drives efforts to find additional molecular biomarkers that would improve the timing of the salvage therapy.
Collapse
Affiliation(s)
- Soňa Argalácsová
- Department of Oncology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Michal Vočka
- Department of Oncology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Otakar Čapoun
- Department of Urology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Lukáš Lambert
- Department of Radiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| |
Collapse
|
6
|
Singh G, Tolkachjov SN, Farberg AS. Incorporation of the 40-Gene Expression Profile (40-GEP) Test to Improve Treatment Decisions in High-Risk Cutaneous Squamous Cell Carcinoma (cSCC) Patients: Case Series and Algorithm. Clin Cosmet Investig Dermatol 2023; 16:925-935. [PMID: 37051586 PMCID: PMC10083143 DOI: 10.2147/ccid.s403330] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/25/2023] [Indexed: 04/08/2023]
Abstract
Cutaneous squamous cell carcinoma (cSCC) has become a significant public health issue due to its rapidly rising incidence and an estimated 1.8 million newly diagnosed cases annually. As with other cancers, treatment decisions for patients with cSCC are based primarily on a patient's risk for poor outcomes. There has been improvement in clinicopathologic factor-based risk assessment approaches, either through informal methods or ever evolving staging approaches. However, these approaches misidentify patients who will eventually have disease progression as low-risk and conversely, over classify patients as high-risk who do not experience relapse. To improve the accuracy of risk assessment for patients with cSCC, the 40-gene expression profile (40-GEP) test has been validated to show statistically significant stratification of a high-risk cSCC patient's risk of nodal or distant metastasis, independent of currently available risk-assessment methods. The 40-GEP test allows for a more accurate classification of metastatic risk for high-risk cSCC patients, with the aim to influence appropriate allocation of clinician time and therapeutic resources to those patients who will most benefit. The objective of this article is to present a treatment algorithm in which clinicians can easily integrate the results of the 40-GEP test into their current treatment approaches to tailor patient care based on individual tumor biology. The following modalities were considered: surveillance imaging, sentinel lymph node biopsy (SLNB), adjuvant radiation therapy (ART), and clinical follow-up. The authors have contributed their own cases for discussion as to how they have seen the beneficial impact of 40-GEP test results in their own practice. Overall, clinicians can identify risk-aligned treatment pathway improvements with the use of the 40-GEP test for challenging to manage, high-risk cSCC patients.
Collapse
Affiliation(s)
| | - Stanislav N Tolkachjov
- Epiphany Dermatology, Dallas, TX, USA
- University of Texas at Southwestern, Dallas, TX, USA
- Baylor University Medical Center, Dallas, TX, USA
- Texas A&M College of Medicine, Dallas, TX, USA
| | - Aaron S Farberg
- Texas A&M College of Medicine, Dallas, TX, USA
- Bare Dermatology, Dallas, TX, USA
- Baylor Scott & White Health System, Dallas, TX, USA
- Correspondence: Aaron S Farberg, Bare Dermatology, 2110 Research Row, Dallas, TX, 75235, USA, Tel +1 847-721-2725, Email
| |
Collapse
|
7
|
Pereira ÉR, Pinheiro LCL, Francelino AL, Miqueloto CA, Guembarovski AFML, de Oliveira KB, Fuganti PE, de Syllos Cólus IM, Guembarovski RL. Tissue immunostaining of candidate prognostic proteins in metastatic and non-metastatic prostate cancer. J Cancer Res Clin Oncol 2023; 149:567-577. [PMID: 36008689 DOI: 10.1007/s00432-022-04274-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 08/06/2022] [Indexed: 12/24/2022]
Abstract
PURPOSE Prostate cancer (PCa) lacks specific markers capable of distinguishing aggressive tumors from those with indolent behavior. Therefore, the aim of this study was to evaluate the immunostaining of candidate proteins (PTEN, AKT, TRPM8, and NKX3.1) through the immunohistochemistry technique (IHC) on patients with metastatic and non-metastatic PCa. METHODS Tissues from 60 patients were divided into three groups categorized according to prognostic parameters: better prognosis (n = 20), worse prognosis (n = 23), and metastatic (n = 17). Immunostaining was analyzed by a pathologist and staining classifications were considered according to signal intensity: (0) no staining, (+) weak, and (++ and +++) intermediate to strong. RESULTS AKT protein was associated (p = 0.012) and correlated (p = 0.014; Tau = - 0.288) with the prognostic groups. The immunostaining for TRPM8 (p = 0.010) and NKX3.1 (p = 0.003) proteins differed between malignant tumor and non-tumoral adjacent tissue as well as for proteins in cellular locations (nucleus and cytoplasm). TRPM8 was independently associated with the ISUP grade ≥ 4 (p = 0.024; OR = 8.373; 95% CI = 1.319-53.164). The NKX3.1 showed positive and predominantly strong immunostaining in all patients in both tumoral and non-tumoral adjacent tissues. All metastatic samples had positive immunostaining, with strong intensity for NKX3.1 (p = 0.021; Tau = - 0.302). In the non-metastatic group, this strong protein staining was not observed in any patients. CONCLUSION This study confirmed that NKX3.1 is highly specific for prostate tissue and indicated that NKX3.1, AKT, and TRPM8 may be candidate markers for prostate cancer prognosis.
Collapse
Affiliation(s)
- Érica Romão Pereira
- Laboratory of Mutagenesis and Oncogenetics, Department of General Biology, Londrina State University, Londrina, PR, Brazil
| | - Laís Capelasso Lucas Pinheiro
- Laboratory of Mutagenesis and Oncogenetics, Department of General Biology, Londrina State University, Londrina, PR, Brazil
| | - Amanda Letícia Francelino
- Laboratory of Mutagenesis and Oncogenetics, Department of General Biology, Londrina State University, Londrina, PR, Brazil
| | - Carlos Alberto Miqueloto
- Laboratory of Extracellular Matrix, Department of General Biology, Londrina State University, Londrina, PR, Brazil
| | | | - Karen Brajão de Oliveira
- Laboratory of Molecular Genetics and Immunology, Department of Pathological Science, Londrina State University, Londrina, PR, Brazil
| | | | - Ilce Mara de Syllos Cólus
- Laboratory of Mutagenesis and Oncogenetics, Department of General Biology, Londrina State University, Londrina, PR, Brazil
| | - Roberta Losi Guembarovski
- Laboratory of Mutagenesis and Oncogenetics, Department of General Biology, Londrina State University, Londrina, PR, Brazil.
| |
Collapse
|
8
|
Ye J, Wu M, He L, Chen P, Liu H, Yang H. Glutathione-S-Transferase p1 Gene Promoter Methylation in Cell-Free DNA as a Diagnostic and Prognostic Tool for Prostate Cancer: A Systematic Review and Meta-Analysis. Int J Endocrinol 2023; 2023:7279243. [PMID: 36747996 PMCID: PMC9899149 DOI: 10.1155/2023/7279243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 01/04/2023] [Accepted: 01/10/2023] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Promoter methylation of glutathione-S-transferase p1 (GSTP1) is related to the occurrence of prostate cancer (PCa), but reports are inconsistent about the accuracy of GSTP1 promoter methylation in PCa diagnosis and prognosis. Therefore, we systematically evaluated the diagnostic and prognostic value of GSTP1 promoter methylation in PCa. METHODS The PubMed, EMBASE, Web of Science, and PMC databases were searched for all relevant studies from the date of inception to November 31, 2021. We compared differences in the incidence of GSTP1 promoter methylation in cfDNA between prostate cancer patients and controls. The odds ratio (OR) and hazard ratio (HR) were used as effect sizes, and the result of each effect size is expressed as a 95% confidence interval (95% CI). RESULTS Our meta-analysis showed that the combined sensitivity and specificity of GSTP1 promoter methylation in cfDNA for the diagnosis of prostate cancer were 0.37 (95% CI = 0.23, 0.53) and 0.97 (95% CI = 0.88, 0.99), respectively. The area under the curve (AUC) with 95% CI was 0.78 (95% CI = 0.75, 0.82). For prognostic variables, hypermethylation of GSTP1 was associated with shorter survival in PCa (HR = 2.57, 95% CI = 1.30, 5.10), with statistical significance in between-study heterogeneity (I 2 = 72%, P=0.006). The results of the subgroup analysis indicated that the heterogeneity of studies may be due to differences in the observed indicators. CONCLUSIONS The results of the meta-analysis substantiate the high specificity of promoter methylation of GSTP1 in cfDNA for the diagnosis of prostate cancer, and it may be used to more precisely evaluate the prognosis of patients with prostate cancer. It may be helpful for the early detection of prostate cancer, but it still must be combined with traditional prostate-specific antigen (PSA) or other methylated genes to accomplish this goal.
Collapse
Affiliation(s)
- Jinghe Ye
- Department of Graduate School, China Medical University, Shenyang, China
- Department of Organ Transplantation Center, General Hospital of Northern Theatre Command, Shenyang, China
| | - Mao Wu
- Department of Graduate School, China Medical University, Shenyang, China
- Department of Urology, General Hospital of Northern Theatre Command, Shenyang, China
| | - Long He
- Department of Organ Transplantation Center, General Hospital of Northern Theatre Command, Shenyang, China
| | - Peng Chen
- Department of Urology, General Hospital of Northern Theatre Command, Shenyang, China
| | - Hongtao Liu
- Department of Urology, General Hospital of Northern Theatre Command, Shenyang, China
| | - Hongwei Yang
- Department of Organ Transplantation Center, General Hospital of Northern Theatre Command, Shenyang, China
| |
Collapse
|
9
|
Buxton AK, Abbasova S, Bevan CL, Leach DA. Liver Microenvironment Response to Prostate Cancer Metastasis and Hormonal Therapy. Cancers (Basel) 2022; 14:6189. [PMID: 36551674 PMCID: PMC9777323 DOI: 10.3390/cancers14246189] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Prostate cancer-associated deaths arise from disease progression and metastasis. Metastasis to the liver is associated with the worst clinical outcomes for prostate cancer patients, and these metastatic tumors can be particularly resistant to the currently widely used chemotherapy and hormonal therapies, such as anti-androgens which block androgen synthesis or directly target the androgen receptor. The incidence of liver metastases is reportedly increasing, with a potential correlation with use of anti-androgen therapies. A key player in prostate cancer progression and therapeutic response is the microenvironment of the tumor(s). This is a dynamic and adaptive collection of cells and proteins, which impart signals and stimuli that can alter biological processes within prostate cancer cells. Investigation in the prostate primary site has demonstrated that cells of the microenvironment are also responsive to hormones and hormonal therapies. In this review, we collate information about what happens when cancer moves to the liver: the types of prostate cancer cells that metastasize there, the response of resident mesenchymal cells of the liver, and how the interactions between the cancer cells and the microenvironment may be altered by hormonal therapy.
Collapse
Affiliation(s)
| | | | - Charlotte L. Bevan
- Division of Cancer, Imperial Centre for Translational & Experimental Medicine, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK
| | - Damien A. Leach
- Division of Cancer, Imperial Centre for Translational & Experimental Medicine, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK
| |
Collapse
|
10
|
Jiang P, Sinha S, Aldape K, Hannenhalli S, Sahinalp C, Ruppin E. Big data in basic and translational cancer research. Nat Rev Cancer 2022; 22:625-639. [PMID: 36064595 PMCID: PMC9443637 DOI: 10.1038/s41568-022-00502-0] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/26/2022] [Indexed: 02/07/2023]
Abstract
Historically, the primary focus of cancer research has been molecular and clinical studies of a few essential pathways and genes. Recent years have seen the rapid accumulation of large-scale cancer omics data catalysed by breakthroughs in high-throughput technologies. This fast data growth has given rise to an evolving concept of 'big data' in cancer, whose analysis demands large computational resources and can potentially bring novel insights into essential questions. Indeed, the combination of big data, bioinformatics and artificial intelligence has led to notable advances in our basic understanding of cancer biology and to translational advancements. Further advances will require a concerted effort among data scientists, clinicians, biologists and policymakers. Here, we review the current state of the art and future challenges for harnessing big data to advance cancer research and treatment.
Collapse
Affiliation(s)
- Peng Jiang
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Sanju Sinha
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kenneth Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sridhar Hannenhalli
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Cenk Sahinalp
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Eytan Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
11
|
Yuan Y, Song J, Wu Q. Aberrant gene expression pattern in the glycolysis-cholesterol synthesis axis is linked with immune infiltration and prognosis in prostate cancer: A bioinformatics analysis. Medicine (Baltimore) 2022; 101:e31416. [PMID: 36316896 PMCID: PMC9622640 DOI: 10.1097/md.0000000000031416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Aberrant lipid metabolism is an early event in tumorigenesis and has been found in a variety of tumor types, especially prostate cancer (PCa). Therefore, We hypothesize that PCa can be stratified into metabolic subgroups based on glycolytic and cholesterogenic related genes, and the different subgroups are closely related to the immune microenvironment. Bioinformatics analysis of genomic, transcriptomic, and clinical data from a comprehensive cohort of PCa patients was performed. Datasets included the Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) dataset, GSE70768, our previously published PCa cohort. The unsupervised cluster analysis was employed to stratify PCa samples based on the expression of metabolic-related genes. Four molecular subtypes were identified, named Glycolytic, Cholesterogenic, Mixed, and Quiescent. Each metabolic subtype has specific features. Among the 4 subtypes, the cholesterogenic subtype exhibited better median survival, whereas patients with high expression of glycolytic genes showed the shortest survival. The mitochondrial pyruvate carriers (MPC) 1 exhibited expression difference between PCa metabolic subgroups, but not for MPCs 2. Glycolytic subtypes had lower immune cell scores, while Cholesterogenic subgroups had higher immune cell scores. Our results demonstrated that metabolic classifications based on specific glycolytic and cholesterol-producing pathways provide new biological insights into previously established subtypes and may guide develop personalized therapies for unique tumor metabolism characteristics.
Collapse
Affiliation(s)
- Yiwen Yuan
- Guizhou Medical University, Guiyang, Guizhou, P.R. China
- Department of Radiology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, P.R. China
| | - Jukun Song
- Department of Oral and Maxillofacial Surgery, the Affiliated Stomatological Hospital of Guizhou Medical University, Guiyang, Guizhou, P.R. China
| | - Qinghua Wu
- Guizhou Medical University, Guiyang, Guizhou, P.R. China
- Department of Radiology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, P.R. China
- *Correspondence: Qinghua Wu, Guizhou Medical University, Guiyang, Guizhou, P.R. China (e-mail: )
| |
Collapse
|
12
|
Pidsley R, Lam D, Qu W, Peters TJ, Luu P, Korbie D, Stirzaker C, Daly RJ, Stricker P, Kench JG, Horvath LG, Clark SJ. Comprehensive methylome sequencing reveals prognostic epigenetic biomarkers for prostate cancer mortality. Clin Transl Med 2022; 12:e1030. [PMID: 36178085 PMCID: PMC9523674 DOI: 10.1002/ctm2.1030] [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: 03/24/2022] [Revised: 08/02/2022] [Accepted: 08/08/2022] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Prostate cancer is a clinically heterogeneous disease with a subset of patients rapidly progressing to lethal-metastatic prostate cancer. Current clinicopathological measures are imperfect predictors of disease progression. Epigenetic changes are amongst the earliest molecular changes in tumourigenesis. To find new prognostic biomarkers to enable earlier intervention and improved outcomes, we performed methylome sequencing of DNA from patients with localised prostate cancer and long-term clinical follow-up. METHODS We used whole-genome bisulphite sequencing (WGBS) to comprehensively map and compare DNA methylation of radical prostatectomy tissue between patients with lethal disease (n = 7) and non-lethal (n = 8) disease (median follow-up 19.5 years). Validation of differentially methylated regions (DMRs) was performed in an independent cohort (n = 185, median follow-up 15 years) using targeted multiplex bisulphite sequencing of candidate regions. Survival was assessed via univariable and multivariable analyses including clinicopathological measures (log-rank and Cox regression models). RESULTS WGBS data analysis identified cancer-specific methylation patterns including CpG island hypermethylation, and hypomethylation of repetitive elements, with increasing disease risk. We identified 1420 DMRs associated with prostate cancer-specific mortality (PCSM), which showed enrichment for gene sets downregulated in prostate cancer and de novo methylated in cancer. Through comparison with public prostate cancer datasets, we refined the DMRs to develop an 18-gene prognostic panel. Applying this panel to an independent cohort, we found significant associations between PCSM and hypermethylation at EPHB3, PARP6, TBX1, MARCH6 and a regulatory element within CACNA2D4. Strikingly in a multivariable model, inclusion of CACNA2D4 methylation was a better predictor of PCSM versus grade alone (Harrell's C-index: 0.779 vs. 0.684). CONCLUSIONS Our study provides detailed methylome maps of non-lethal and lethal prostate cancer and identifies novel genic regions that distinguish these patient groups. Inclusion of our DNA methylation biomarkers with existing clinicopathological measures improves prognostic models of prostate cancer mortality, and holds promise for clinical application.
Collapse
Affiliation(s)
- Ruth Pidsley
- Garvan Institute of Medical ResearchSydneyNew South WalesAustralia,School of Clinical MedicineSt Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW SydneySydneyNew South WalesAustralia
| | - Dilys Lam
- Garvan Institute of Medical ResearchSydneyNew South WalesAustralia,Present address:
School of Molecular Sciences, The University of Western Australia, Crawley, Western Australia 6009, Australia,Present address:
Harry Perkins Institute of Medical Research, Nedlands, Western Australia 6009, Australia
| | - Wenjia Qu
- Garvan Institute of Medical ResearchSydneyNew South WalesAustralia
| | - Timothy J. Peters
- Garvan Institute of Medical ResearchSydneyNew South WalesAustralia,School of Clinical MedicineSt Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW SydneySydneyNew South WalesAustralia
| | - Phuc‐Loi Luu
- Garvan Institute of Medical ResearchSydneyNew South WalesAustralia,School of Clinical MedicineSt Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW SydneySydneyNew South WalesAustralia
| | - Darren Korbie
- Centre for Personalised NanomedicineAustralian Institute for Bioengineering and NanotechnologyThe University of QueenslandSt. LuciaQueenslandAustralia
| | - Clare Stirzaker
- Garvan Institute of Medical ResearchSydneyNew South WalesAustralia,School of Clinical MedicineSt Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW SydneySydneyNew South WalesAustralia
| | - Roger J. Daly
- Cancer Research Program and Department of Biochemistry and Molecular BiologyBiomedicine Discovery InstituteMonash UniversityClaytonVictoriaAustralia
| | - Phillip Stricker
- Garvan Institute of Medical ResearchSydneyNew South WalesAustralia,School of Clinical MedicineSt Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW SydneySydneyNew South WalesAustralia,Department of UrologySt. Vincent's Prostate Cancer CentreSydneyNew South WalesAustralia
| | - James G. Kench
- Garvan Institute of Medical ResearchSydneyNew South WalesAustralia,Department of Tissue PathologyNSW Health PathologyRoyal Prince Alfred HospitalCamperdownSydneyNew South WalesAustralia
| | - Lisa G. Horvath
- Garvan Institute of Medical ResearchSydneyNew South WalesAustralia,School of Clinical MedicineSt Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW SydneySydneyNew South WalesAustralia,Chris O'Brien Lifehouse, CamperdownSydneyNew South WalesAustralia,University of SydneySydneyNew South WalesAustralia
| | - Susan J. Clark
- Garvan Institute of Medical ResearchSydneyNew South WalesAustralia,School of Clinical MedicineSt Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW SydneySydneyNew South WalesAustralia
| |
Collapse
|
13
|
Maia R, Santos GAD, Reis S, Viana NI, Pimenta R, Guimarães VR, Recuero S, Romão P, Leite KRM, Srougi M, Passerotti CC. Can we use Ki67 expression to predict prostate cancer aggressiveness? Rev Col Bras Cir 2022; 49:e20223200. [PMID: 35792806 PMCID: PMC10578861 DOI: 10.1590/0100-6991e-20223200-en] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 04/06/2022] [Indexed: 11/22/2022] Open
Abstract
INTRODUCTION specialists have an urge for biomarkers that can discriminate indolent prostate cancer from aggressive tumors. Ki67 is a proliferation marker, and its expression is associated with the aggressiveness of several cancers. OBJECTIVE analyze the expression of Ki67 in prostate cancer samples correlating with the aggressiveness of the disease. METHODS Ki67 mRNA levels were determined utilizing data from a TCGA cohort (Tumor(n)=492 and control(n)=52). The protein expression was determined on 94 biopsies from patients by immunohistochemical assay. RESULTS in mRNA, the Ki67 upregulation is associated with cancer tissue (p<0.0001) and worst disease-free survival (p=0.035). The protein upregulation is associated with increase of the ISUP score (p<0.0001), cancer stage (p=0.05), biochemical recurrence (p=0.0006) and metastasis (p<0.0001). We also show a positive correlation between Ki67 expression and ISUP score (r=0.5112, p<0.0001) and disease risk stratification (r=0.3388, p=0.0009). Ki67 expression is a factor independently associated with biochemical recurrence (p=0.002) and metastasis (p<0.0001). Finally, the patients with high Ki67expression shows better survival regarding biochemical recurrence (p=0.008) and metastasis (p=0.056). Patients with high Ki67 expression are 2.62 times more likely to develop biochemical recurrence (p=0.036). CONCLUSION Ki67 upregulation is associated with prostate cancer aggressiveness.
Collapse
Affiliation(s)
- Ronaldo Maia
- - Hospital Alemão Oswaldo Cruz, Center for Robotic Surgery - São Paulo - SP - Brasil
| | - Gabriel Arantes Dos Santos
- - Faculdade de Medicina da Universidade de São Paulo (FMUSP), Urologia - São Paulo - SP - Brasil
- - D'Or Institute for Research and Education (IDOR) - São Paulo - SP - Brasil
| | - Sabrina Reis
- - Faculdade de Medicina da Universidade de São Paulo (FMUSP), Urologia - São Paulo - SP - Brasil
- - Hospital Moriah - São Paulo - SP - Brasil
- - Universidade do Estado de Minas Gerais (UEMG) - Passos - MG - Brasil
| | - Nayara I Viana
- - Hospital Alemão Oswaldo Cruz, Center for Robotic Surgery - São Paulo - SP - Brasil
| | - Ruan Pimenta
- - Faculdade de Medicina da Universidade de São Paulo (FMUSP), Urologia - São Paulo - SP - Brasil
- - D'Or Institute for Research and Education (IDOR) - São Paulo - SP - Brasil
| | - Vanessa R Guimarães
- - Faculdade de Medicina da Universidade de São Paulo (FMUSP), Urologia - São Paulo - SP - Brasil
| | - Saulo Recuero
- - Faculdade de Medicina da Universidade de São Paulo (FMUSP), Urologia - São Paulo - SP - Brasil
| | - Poliana Romão
- - Faculdade de Medicina da Universidade de São Paulo (FMUSP), Urologia - São Paulo - SP - Brasil
| | | | - Miguel Srougi
- - Faculdade de Medicina da Universidade de São Paulo (FMUSP), Urologia - São Paulo - SP - Brasil
- - D'Or Institute for Research and Education (IDOR) - São Paulo - SP - Brasil
| | | |
Collapse
|
14
|
Glicksman R, Kishan A, Quon H, Shabsovich D, Juarez J, Jiang T, Steinberg M, Zhang L, Loblaw A. Absolute Percentage of Pattern 4 Disease as a Prognostic Measure for Intermediate-risk Prostate Cancer Treated with Stereotactic Body Radiotherapy. Clin Oncol (R Coll Radiol) 2022; 34:581-588. [DOI: 10.1016/j.clon.2022.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 04/13/2022] [Accepted: 05/05/2022] [Indexed: 11/25/2022]
|
15
|
Rijstenberg LL, Hansum T, Kweldam CF, Kümmerlin IP, Remmers S, Roobol MJ, van Leenders GJLH. Large and small cribriform architecture have similar adverse clinical outcome on prostate cancer biopsies. Histopathology 2022; 80:1041-1049. [PMID: 35384019 PMCID: PMC9321809 DOI: 10.1111/his.14658] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 03/08/2022] [Accepted: 03/31/2022] [Indexed: 11/29/2022]
Abstract
Aims Invasive cribriform and intraductal carcinoma (IDC) are associated with adverse outcome in prostate cancer patients, with the large cribriform pattern having the worst outcome in radical prostatectomies. Our objective was to determine the impact of the large and small cribriform patterns in prostate cancer biopsies. Methods and results Pathological revision was carried out on biopsies of 1887 patients from the European Randomised Study of Screening for Prostate Cancer. The large cribriform pattern was defined as having at least twice the size of adjacent benign glands. The median follow‐up time was 13.4 years. Hazard ratios for metastasis‐free survival (MFS) and disease‐specific survival (DSS) were calculated using Cox proportional hazards regression. Any cribriform pattern was found in 280 of 1887 men: 1.1% IDC in grade group (GG) 1, 18.2% in GG2, 57.1% in GG3, 55.4% in GG4 and 59.3% in GG5; the large cribriform pattern was present in 0, 0.5, 9.8, 18.1 and 17.3%, respectively. In multivariable analyses, small and large cribriform patterns were both (P < 0.005) associated with worse MFS [small: hazard ratio (HR) = 3.04, 95% confidence interval (CI) = 1.93–4.78; large: HR = 3.17, 95% CI = 1.68–5.99] and DSS (small: HR = 4.07, 95% CI = 2.51–6.62; large: HR = 4.13, 95% CI = 2.14–7.98). Patients with the large cribriform pattern did not have worse MFS (P = 0.77) or DSS (P = 0.96) than those with the small cribriform pattern. Conclusions Both small and large cribriform patterns are associated with worse MFS and DSS in prostate cancer biopsies. Patients with the large cribriform pattern on biopsy have a similar adverse outcome as those with the small cribriform pattern.
Collapse
Affiliation(s)
- L Lucia Rijstenberg
- Department of Pathology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Tim Hansum
- Department of Pathology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Charlotte F Kweldam
- Department of Pathology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands.,Department of Pathology, Maasstad Hospital, Rotterdam, the Netherlands
| | - Intan P Kümmerlin
- Department of Pathology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Sebastiaan Remmers
- Department of Urology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Monique J Roobol
- Department of Urology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | | |
Collapse
|
16
|
Macrini S, Francesconi S, Caprera C, Lancia D, Corsi M, Gunnellini M, Rocchi A, Pireddu A, Marziani F, Mosillo C, Calandrella ML, Caserta C, Giannarelli D, Guida A, Ascani S, Bracarda S. Looking for a Simplified Diagnostic Model to Identify Potentially Lethal Cases of Prostate Cancer at Initial Diagnosis: An ImGO Pilot Study. Cancers (Basel) 2022; 14:1542. [PMID: 35326693 PMCID: PMC8946832 DOI: 10.3390/cancers14061542] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/11/2022] [Accepted: 03/12/2022] [Indexed: 12/12/2022] Open
Abstract
The recurrent genetic anomalies used to classify prostate cancer (PC) into distinct molecular subtypes have limited relevance for clinical practice. In consideration of WHO 2016 histological classification, which includes the introduction of Gleason Score 4 for patients with cribriform component and the definition of intraductal carcinoma as a new entity, a retrospective pilot study was conducted to investigate, by histological review, if there were any variations of Gleason Score and the incidence of intraductal carcinoma and cribriform pattern, intended as "phenotypic" markers of potentially lethal PC, among metastatic castration-sensitive PC (mCSPC) and metastatic castration-resistant PC (mCRPC) samples. Potentially predictive factors were also assessed. Among 125 cases, a variation in the Gleason Score was reported in 26% of cases. A cribriform (36%) or intraductal (2%) pattern was reported in a higher percentage. Of them, a primary Gleason pattern 4 was reported in 80% of cases. All patients with intraductal carcinoma present a BRCA2 mutation, also found in 80% of cases with a cribriform pattern. This pilot study documented some hypothesis-generating data, as the evaluation of de novo mCSPC and mCRPC as phenotypic/biologic model to be translated in clinical practice. A cribriform pattern/intraductal carcinoma might be a marker of potentially lethal PC. The high incidence of TP53 and BRCA2 mutations in de novo mCSPC may also have a therapeutic implication.
Collapse
Affiliation(s)
- Serena Macrini
- Medical and Translational Oncology Unit, Department of Oncology, Azienda Ospedaliera Santa Maria, 05100 Terni, Italy; (S.M.); (C.M.); (M.L.C.); (C.C.); (A.G.)
| | - Simona Francesconi
- Pathology Unit, Azienda Ospedaliera Santa Maria Terni, University of Perugia, 06129 Terni, Italy; (S.F.); (C.C.); (D.L.); (M.C.); (S.A.)
| | - Cecilia Caprera
- Pathology Unit, Azienda Ospedaliera Santa Maria Terni, University of Perugia, 06129 Terni, Italy; (S.F.); (C.C.); (D.L.); (M.C.); (S.A.)
| | - Daniela Lancia
- Pathology Unit, Azienda Ospedaliera Santa Maria Terni, University of Perugia, 06129 Terni, Italy; (S.F.); (C.C.); (D.L.); (M.C.); (S.A.)
| | - Matteo Corsi
- Pathology Unit, Azienda Ospedaliera Santa Maria Terni, University of Perugia, 06129 Terni, Italy; (S.F.); (C.C.); (D.L.); (M.C.); (S.A.)
| | - Marco Gunnellini
- Medical Oncology Unit, Department of Oncology, Gubbio-Gualdo Tadino Hospital, 06024 Branca, Italy;
| | - Andrea Rocchi
- Medical Oncology Unit, Department of Medicine, San Giovanni Battista Hospital, 06034 Foligno, Italy;
| | - Anjuta Pireddu
- Division of Pathology, Città di Castello Hospital, 06012 Città di Castello, Italy;
| | - Fiovo Marziani
- Pathology Unit, Department of Clinical Pathology, San Giovanni Battista Hospital, 06034 Foligno, Italy;
| | - Claudia Mosillo
- Medical and Translational Oncology Unit, Department of Oncology, Azienda Ospedaliera Santa Maria, 05100 Terni, Italy; (S.M.); (C.M.); (M.L.C.); (C.C.); (A.G.)
| | - Maria Letizia Calandrella
- Medical and Translational Oncology Unit, Department of Oncology, Azienda Ospedaliera Santa Maria, 05100 Terni, Italy; (S.M.); (C.M.); (M.L.C.); (C.C.); (A.G.)
| | - Claudia Caserta
- Medical and Translational Oncology Unit, Department of Oncology, Azienda Ospedaliera Santa Maria, 05100 Terni, Italy; (S.M.); (C.M.); (M.L.C.); (C.C.); (A.G.)
| | - Diana Giannarelli
- Biostatistical Unit, Regina Elena National Cancer Institute, IRCCS, 00168 Rome, Italy;
| | - Annalisa Guida
- Medical and Translational Oncology Unit, Department of Oncology, Azienda Ospedaliera Santa Maria, 05100 Terni, Italy; (S.M.); (C.M.); (M.L.C.); (C.C.); (A.G.)
| | - Stefano Ascani
- Pathology Unit, Azienda Ospedaliera Santa Maria Terni, University of Perugia, 06129 Terni, Italy; (S.F.); (C.C.); (D.L.); (M.C.); (S.A.)
| | - Sergio Bracarda
- Medical and Translational Oncology Unit, Department of Oncology, Azienda Ospedaliera Santa Maria, 05100 Terni, Italy; (S.M.); (C.M.); (M.L.C.); (C.C.); (A.G.)
| |
Collapse
|
17
|
Packeiser EM, Taher L, Kong W, Ernst M, Beck J, Hewicker-Trautwein M, Brenig B, Schütz E, Murua Escobar H, Nolte I. RNA-seq of nine canine prostate cancer cell lines reveals diverse therapeutic target signatures. Cancer Cell Int 2022; 22:54. [PMID: 35109825 PMCID: PMC8812184 DOI: 10.1186/s12935-021-02422-9] [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: 05/01/2021] [Accepted: 12/21/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Canine prostate adenocarcinoma (PAC) and transitional cell carcinoma (TCC) are typically characterized by metastasis and chemoresistance. Cell lines are important model systems for developing new therapeutic strategies. However, as they adapt to culturing conditions and undergo clonal selection, they can diverge from the tissue from which they were originally derived. Therefore, a comprehensive characterization of cell lines and their original tissues is paramount. METHODS This study compared the transcriptomes of nine canine cell lines derived from PAC, PAC metastasis and TCC to their respective original primary tumor or metastasis tissues. Special interests were laid on cell culture-related differences, epithelial to mesenchymal transition (EMT), the prostate and bladder cancer pathways, therapeutic targets in the PI3K-AKT signaling pathway and genes correlated with chemoresistance towards doxorubicin and carboplatin. RESULTS Independent analyses for PAC, PAC metastasis and TCC revealed 1743, 3941 and 463 genes, respectively, differentially expressed in the cell lines relative to their original tissues (DEGs). While genes associated with tumor microenvironment were mostly downregulated in the cell lines, patient-specific EMT features were conserved. Furthermore, examination of the prostate and bladder cancer pathways revealed extensive concordance between cell lines and tissues. Interestingly, all cell lines preserved downstream PI3K-AKT signaling, but each featured a unique therapeutic target signature. Additionally, resistance towards doxorubicin was associated with G2/M cell cycle transition and cell membrane biosynthesis, while carboplatin resistance correlated with histone, m- and tRNA processing. CONCLUSION Comparative whole-transcriptome profiling of cell lines and their original tissues identifies models with conserved therapeutic target expression. Moreover, it is useful for selecting suitable negative controls, i.e., cell lines lacking therapeutic target expression, increasing the transfer efficiency from in vitro to primary neoplasias for new therapeutic protocols. In summary, the dataset presented here constitutes a rich resource for canine prostate and bladder cancer research.
Collapse
Affiliation(s)
- Eva-Maria Packeiser
- Small Animal Clinic, University of Veterinary Medicine Hannover, Hannover, Germany
- Department of Medicine, Clinic III, Hematology, Oncology and Palliative Medicine, University Medical Center Rostock, Rostock, Germany
| | - Leila Taher
- Institute of Biomedical Informatics, Graz University of Technology, Graz, Austria
- Division of Bioinformatics, Department of Biology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, 18057, Rostock, Germany
| | - Weibo Kong
- Small Animal Clinic, University of Veterinary Medicine Hannover, Hannover, Germany
- Department of Medicine, Clinic III, Hematology, Oncology and Palliative Medicine, University Medical Center Rostock, Rostock, Germany
- Institute of Muscle Biology and Growth, Research Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany
| | - Mathias Ernst
- Division of Bioinformatics, Department of Biology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | | | | | - Bertram Brenig
- University of Göttingen, Institute of Veterinary Medicine, Göttingen, Germany
| | | | - Hugo Murua Escobar
- Small Animal Clinic, University of Veterinary Medicine Hannover, Hannover, Germany.
- Department of Medicine, Clinic III, Hematology, Oncology and Palliative Medicine, University Medical Center Rostock, Rostock, Germany.
- Comprehensive Cancer Center Mecklenburg-Vorpommern (CCC-MV), Campus Rostock, University of Rostock, 18057, Rostock, Germany.
| | - Ingo Nolte
- Small Animal Clinic, University of Veterinary Medicine Hannover, Hannover, Germany.
| |
Collapse
|
18
|
Current Methods and Caveats to Risk Factor Assessment in Cutaneous Squamous Cell Carcinoma (cSCC): A Narrative Review. Dermatol Ther (Heidelb) 2022; 12:267-284. [PMID: 34994967 PMCID: PMC8850485 DOI: 10.1007/s13555-021-00673-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Indexed: 01/21/2023] Open
Abstract
Cutaneous squamous cell carcinoma (cSCC) is the second most common form of skin cancer, and the number of deaths due to cSCC is estimated to be greater than the number attributed to melanoma. While the majority of cSCC tumors are resectable with clear margins by standard excision practices, some lesions exhibit high-risk factors for which there is evidence of their association with recurrence, metastasis, and disease-specific death. The most commonly used staging systems and guidelines in the USA for cSCC are based on these clinical and pathologic high-risk factors; however, these are limited in their ability to predict adverse events, thus posing a challenge for implementing risk-directed patient management. Since the development of local recurrence and/or metastasis has a profound impact on the survival of patients with cSCC, accurate identification of patients at high risk for poor outcomes is critical, potentially allowing for early and appropriate adjuvant therapy. This review summarizes the current cSCC literature with a focus on how differing clinical assessments within each of the five selected risk factors (perineural invasion, differentiation, depth of invasion, size, and location) can influence the evaluation of patient outcomes, along with summarizing the utility of staging and guidelines, and highlighting the potential for molecular tools to improve upon cSCC risk assessment.
Collapse
|
19
|
Şeref C, Acar Ö, Kılıç M, Vural M, Sağlıcan Y, Saraç H, Coşkun B, İnce Ü, Esen T, Lack NA. Histologically benign PI-RADS 4 and 5 lesions contain cancer-associated epigenetic alterations. Prostate 2022; 82:145-153. [PMID: 34672371 DOI: 10.1002/pros.24255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/31/2021] [Accepted: 09/29/2021] [Indexed: 11/07/2022]
Abstract
BACKGROUND The detection rate of clinically significant prostate cancer has improved with the use of multiparametric magnetic resonance imaging (mpMRI). Yet, even with MRI-guided biopsy 15%-35% of high-risk lesions (Prostate Imaging-Reporting and Data System [PI-RADS] 4 and 5) are histologically benign. It is unclear if these false positives are due to diagnostic/sampling errors or pathophysiological alterations. To better understand this, we tested histologically benign PI-RAD 4 and 5 lesions for common malignant epigenetic alterations. MATERIALS AND METHODS MRI-guided in-bore biopsy samples were collected from 45 patients with PI-RADS 4 (n = 31) or 5 (n = 14) lesions. Patients had a median clinical follow-up of 3.8 years. High-risk mpMRI patients were grouped based on their histology into biopsy positive for tumor (BPT; n = 28) or biopsy negative for tumor (BNT; n = 17). From these biopsy samples, DNA methylation of well-known tumor suppressor genes (APC, GSTP1, and RARβ2) was quantified. RESULTS Similar to previous work we observed high rates of promoter methylation at GSTP1 (92.7%), RARβ2 (57.3%), and APC (37.8%) in malignant BPT samples but no methylation in benign TURP chips. Interestingly, similar to the malignant samples the BNT biopsies also had increased methylation at the promoter of GSTP1 (78.8%) and RARβ2 (34.6%). However, despite these epigenetic alterations none of these BNT patients developed prostate cancer, and those who underwent repeat mpMRI (n = 8) demonstrated either radiological regression or stability. CONCLUSIONS Histologically benign PI-RADS 4 and 5 lesions harbor prostate cancer-associated epigenetic alterations.
Collapse
Affiliation(s)
- Ceren Şeref
- Department of Health Sciences, Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Ömer Acar
- Department of Urology, Koc University School of Medicine, Istanbul, Turkey
| | - Mert Kılıç
- Department of Urology, VKF American Hospital, Istanbul, Turkey
| | - Metin Vural
- Department of Radiology, VKF American Hospital, Istanbul, Turkey
| | - Yeşim Sağlıcan
- Department of Pathology, Acıbadem University School of Medicine, Istanbul, Turkey
| | - Hilal Saraç
- Department of Health Sciences, Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Bilgen Coşkun
- Department of Radiology, VKF American Hospital, Istanbul, Turkey
| | - Ümit İnce
- Department of Pathology, Acıbadem University School of Medicine, Istanbul, Turkey
| | - Tarık Esen
- Department of Urology, Koc University School of Medicine, Istanbul, Turkey
- Department of Urology, VKF American Hospital, Istanbul, Turkey
| | - Nathan A Lack
- Department of Health Sciences, Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
- Department of Medical Pharmacology, Koç University School of Medicine, Istanbul, Turkey
- Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| |
Collapse
|
20
|
MAIA RONALDO, SANTOS GABRIELARANTESDOS, REIS SABRINA, VIANA NAYARAI, PIMENTA RUAN, GUIMARÃES VANESSAR, RECUERO SAULO, ROMÃO POLIANA, LEITE KATIARAMOSMOREIRA, SROUGI MIGUEL, PASSEROTTI CARLOCARMARGO. Podemos usar a expressão de Ki67 para prever a agressividade do câncer de próstata? Rev Col Bras Cir 2022. [DOI: 10.1590/0100-6991e-20223200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
RESUMO Introdução: especialistas precisam biomarcadores que podem discriminar o câncer de próstata indolente de tumores agressivos. Ki67 é um marcador de proliferação, e sua expressão está associada à agressividade de vários tumores. Objetivo: analisar a expressão do Ki67 em amostras de câncer de próstata correlacionando com a agressividade da doença. Métodos: os níveis de mRNA de Ki67 foram determinados utilizando dados de uma coorte de TCGA (Tumor(n)=492 e controle(n)=52). A expressão da proteína foi determinada em 94 biópsias de pacientes por ensaio imuno-histoquímica. Resultados: no mRNA, a superexpressão Ki67 está associada ao tecido canceroso (p<0,0001) e à pior sobrevida livre de doença (p=0,035). A superexpressão proteica está associada ao aumento do escore ISUP (p<0,0001), estágio de câncer (p=0,05), recorrência bioquímica (p=0,0006) e metástase (p<0,0001). Também mostramos uma correlação positiva entre a expressão Ki67 e o escore ISUP (r=0,5112, p<0,0001) e a estratificação de risco de doença (r=0,3388, p=0,0009). A expressão Ki67 é um fator independentemente associado à recorrência bioquímica (p=0,002) e metástase (p<0,0001). Finalmente, os pacientes com alta expressão de Ki67 expression mostram melhor sobrevivência em relação à recorrência bioquímica (p=0,008) e metástase (p=0,056). Os pacientes com alta expressão de Ki67 são 2,62 vezes mais propensos a desenvolver recorrência bioquímica (p=0,036). Conclusão: a superexpressão Ki67 está associada à agressividade do câncer de próstata.
Collapse
Affiliation(s)
| | | | - SABRINA REIS
- Universidade de São Paulo, Brazil; Hospital Moriah, Brasil; Universidade do Estado de Minas Gerais, Brazil
| | | | - RUAN PIMENTA
- Universidade de São Paulo, Brazil; D’Or Institute for Research and Education, Brasil
| | | | | | | | | | - MIGUEL SROUGI
- Universidade de São Paulo, Brazil; D’Or Institute for Research and Education, Brasil
| | | |
Collapse
|
21
|
Kristiansen G, Schmid M. Application of computer-generated images to train pattern recognition used in semiquantitative immunohistochemistry scoring. APMIS 2021; 130:26-33. [PMID: 34748225 DOI: 10.1111/apm.13188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
This study aimed to clarify whether the pattern recognition involved in scoring proliferation fractions can be trained by abstract computerized images of virtual tissues. Twenty computer-generated images with randomly distributed blue or red dots were scored by 12 probands (all co-workers or collaborators of the Institute of Pathology, University of Bonn). Afterward, the probands underwent a training phase during which they received an immediate feedback on the actual rate of positivity after each image. Finally, the initial testing series was rescored. In a second round with 15 different probands, 20 Ki-67 immunohistochemistry images of tonsil tissue were scored, followed by the same training phase with computer-generated images, before the immunohistochemistry slides were scored again. Paired t-tests were used to compare the differences in mean rates pre- and post-training. Concerning computerized images, untrained probands scored the percentages of positive dots with a mean deviation from the true rates of 8.2%. Following training, the same testing series was scored significantly better with a mean deviation of 4.9% (mean improvement 3.3%, p < 0.001). Scoring real immunohistochemistry slides, the training with computerized images also improved correct estimations, albeit to a lesser degree (mean improvement 1%, p = 0.03). Abstract computerized images of virtual tissues may be a useful tool to train and improve the accuracy of pattern recognition involved in semiquantitative scoring of immunohistochemistry slides. As a side results, this study highlights the value of computer-generated images to verify the performance of image-analysis software.
Collapse
Affiliation(s)
| | - Matthias Schmid
- Institute of Medical Biometry, Informatics and Epidemiology (IMBIE), University Hospital Bonn, Bonn, Germany
| |
Collapse
|
22
|
Thiemeyer H, Taher L, Schille JT, Packeiser EM, Harder LK, Hewicker-Trautwein M, Brenig B, Schütz E, Beck J, Nolte I, Murua Escobar H. An RNA-Seq-Based Framework for Characterizing Canine Prostate Cancer and Prioritizing Clinically Relevant Biomarker Candidate Genes. Int J Mol Sci 2021; 22:11481. [PMID: 34768937 PMCID: PMC8584104 DOI: 10.3390/ijms222111481] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/16/2021] [Accepted: 10/16/2021] [Indexed: 01/01/2023] Open
Abstract
Prostate cancer (PCa) in dogs is a highly malignant disease akin to its human counterpart. In contrast to the situation in humans, multi-gene approaches facilitating risk stratification of canine PCa are barely established. The aims of this study were the characterization of the transcriptional landscape of canine PCa and the identification of diagnostic, prognostic and/or therapeutic biomarkers through a multi-step screening approach. RNA-Sequencing of ten malignant tissues and fine-needle aspirations (FNA), and 14 nonmalignant tissues and FNAs was performed to find differentially expressed genes (DEGs) and deregulated pathways. The 4098 observed DEGs were involved in 49 pathways. These 49 pathways could be grouped into five superpathways summarizing the hallmarks of canine PCa: (i) inflammatory response and cytokines; (ii) regulation of the immune system and cell death; (iii) cell surface and PI3K signaling; (iv) cell cycle; and (v) phagosome and autophagy. Among the highly deregulated, moderately to strongly expressed DEGs that were members of one or more superpathways, 169 DEGs were listed in relevant databases and/or the literature and included members of the PCa pathway, oncogenes, prostate-specific genes, and druggable genes. These genes are novel and promising candidate diagnostic, prognostic and/or therapeutic canine PCa biomarkers.
Collapse
Affiliation(s)
- Heike Thiemeyer
- Small Animal Clinic, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany; (H.T.); (J.T.S.); (E.-M.P.); (L.K.H.); (I.N.)
- Department of Hematology/Oncology/Palliative Care, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Leila Taher
- Institute of Biomedical Informatics, Graz University of Technology, 8010 Graz, Austria;
| | - Jan Torben Schille
- Small Animal Clinic, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany; (H.T.); (J.T.S.); (E.-M.P.); (L.K.H.); (I.N.)
- Department of Hematology/Oncology/Palliative Care, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Eva-Maria Packeiser
- Small Animal Clinic, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany; (H.T.); (J.T.S.); (E.-M.P.); (L.K.H.); (I.N.)
- Department of Hematology/Oncology/Palliative Care, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Lisa K. Harder
- Small Animal Clinic, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany; (H.T.); (J.T.S.); (E.-M.P.); (L.K.H.); (I.N.)
| | - Marion Hewicker-Trautwein
- Institute of Pathology, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany;
| | - Bertram Brenig
- Institute of Veterinary Medicine, University of Göttingen, 37077 Göttingen, Germany;
| | - Ekkehard Schütz
- Chronix Biomedical GmbH, 37079 Göttingen, Germany; (E.S.); (J.B.)
| | - Julia Beck
- Chronix Biomedical GmbH, 37079 Göttingen, Germany; (E.S.); (J.B.)
| | - Ingo Nolte
- Small Animal Clinic, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany; (H.T.); (J.T.S.); (E.-M.P.); (L.K.H.); (I.N.)
| | - Hugo Murua Escobar
- Small Animal Clinic, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany; (H.T.); (J.T.S.); (E.-M.P.); (L.K.H.); (I.N.)
- Department of Hematology/Oncology/Palliative Care, Rostock University Medical Centre, 18057 Rostock, Germany
- Comprehensive Cancer Center Mecklenburg-Vorpommern (CCC-MV), Campus Rostock, University of Rostock, 18057 Rostock, Germany
| |
Collapse
|
23
|
Giunta EF, Annaratone L, Bollito E, Porpiglia F, Cereda M, Banna GL, Mosca A, Marchiò C, Rescigno P. Molecular Characterization of Prostate Cancers in the Precision Medicine Era. Cancers (Basel) 2021; 13:4771. [PMID: 34638258 PMCID: PMC8507555 DOI: 10.3390/cancers13194771] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/16/2021] [Accepted: 09/20/2021] [Indexed: 12/31/2022] Open
Abstract
Prostate cancer (PCa) therapy has been recently revolutionized by the approval of new therapeutic agents in the metastatic setting. However, the optimal therapeutic strategy in such patients should be individualized in the light of prognostic and predictive molecular factors, which have been recently studied: androgen receptor (AR) alterations, PTEN-PI3K-AKT pathway deregulation, homologous recombination deficiency (HRD), mismatch repair deficiency (MMRd), and tumor microenvironment (TME) modifications. In this review, we highlighted the clinical impact of prognostic and predictive molecular factors in PCa patients' outcomes, identifying biologically distinct subtypes. We further analyzed the relevant methods to detect these factors, both on tissue, i.e., immunohistochemistry (IHC) and molecular tests, and blood, i.e., analysis of circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA). Moreover, we discussed the main pros and cons of such techniques, depicting their present and future roles in PCa management, throughout the precision medicine era.
Collapse
Affiliation(s)
- Emilio Francesco Giunta
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80131 Naples, Italy;
| | - Laura Annaratone
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (L.A.); (C.M.)
- Pathology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060 Turin, Italy
| | - Enrico Bollito
- Department of Pathology, University of Turin, San Luigi Gonzaga Hospital, Orbassano, 10043 Turin, Italy;
| | - Francesco Porpiglia
- Department of Urology, University of Turin, San Luigi Gonzaga Hospital, Orbassano, 10043 Turin, Italy;
| | - Matteo Cereda
- Cancer Genomics and Bioinformatics Unit, IIGM-Italian Institute for Genomic Medicine, c/o IRCCS Candiolo, 10060 Turin, Italy;
- Candiolo Cancer Institute, FPO—IRCCS, Str. Prov.le 142, km 3.95, 10060 Candiolo, Italy
| | - Giuseppe Luigi Banna
- Department of Oncology, Portsmouth Hospitals University NHS Trust, Portsmouth PO2 8QD, UK;
| | - Alessandra Mosca
- Multidisciplinary Outpatient Oncology Clinic, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060 Turin, Italy;
| | - Caterina Marchiò
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (L.A.); (C.M.)
- Pathology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060 Turin, Italy
| | - Pasquale Rescigno
- Interdisciplinary Group for Translational Research and Clinical Trials, Urological Cancers (GIRT-Uro), Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060 Turin, Italy
| |
Collapse
|
24
|
Newman JG, Hall MA, Kurley SJ, Cook RW, Farberg AS, Geiger JL, Koyfman SA. Adjuvant therapy for high-risk cutaneous squamous cell carcinoma: 10-year review. Head Neck 2021; 43:2822-2843. [PMID: 34096664 PMCID: PMC8453797 DOI: 10.1002/hed.26767] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 04/29/2021] [Accepted: 05/18/2021] [Indexed: 12/16/2022] Open
Abstract
Standard of care for high-risk cutaneous squamous cell carcinoma (cSCC) is surgical excision of the primary lesion with clear margins when possible, and additional resection of positive margins when feasible. Even with negative margins, certain high-risk factors warrant consideration of adjuvant therapy. However, which patients might benefit from adjuvant therapy is unclear, and supporting evidence is conflicting and limited to mostly small retrospective cohorts. Here, we review literature from the last decade regarding adjuvant radiation therapy and systemic therapy in high-risk cSCC, including recent and current trials and the role of immune checkpoint inhibitors. We demonstrate evidence gaps in adjuvant therapy for high-risk cSCC and the need for prognostic tools, such as gene expression profiling, to guide patient selection. More large-cohort clinical studies are needed for collecting high-quality, evidence-based data for determining which patients with high-risk cSCC may benefit from adjuvant therapy and which therapy is most appropriate for patient management.
Collapse
Affiliation(s)
- Jason G. Newman
- Department of Otorhinolaryngology – Head and Neck SurgeryUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Mary A. Hall
- Research and Development, Castle Biosciences, Inc.FriendswoodTexasUSA
| | - Sarah J. Kurley
- Research and Development, Castle Biosciences, Inc.FriendswoodTexasUSA
| | - Robert W. Cook
- Research and Development, Castle Biosciences, Inc.FriendswoodTexasUSA
| | - Aaron S. Farberg
- Section of DermatologyBaylor University Medical CenterDallasTexasUSA
| | - Jessica L. Geiger
- Department of Hematology and Medical OncologyCleveland ClinicClevelandOhioUSA
| | | |
Collapse
|
25
|
Rounds L, Nagle RB, Muranyi A, Jandova J, Gill S, Vela E, Wondrak GT. Glyoxalase 1 Expression as a Novel Diagnostic Marker of High-Grade Prostatic Intraepithelial Neoplasia in Prostate Cancer. Cancers (Basel) 2021; 13:3608. [PMID: 34298821 PMCID: PMC8304603 DOI: 10.3390/cancers13143608] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/12/2021] [Accepted: 07/15/2021] [Indexed: 12/03/2022] Open
Abstract
Glyoxalase 1 (GLO1) is an enzyme involved in the detoxification of methylglyoxal (MG), a reactive oncometabolite formed in the context of energy metabolism as a result of high glycolytic flux. Prior clinical evidence has documented GLO1 upregulation in various tumor types including prostate cancer (PCa). However, GLO1 expression has not been explored in the context of PCa progression with a focus on high-grade prostatic intraepithelial neoplasia (HGPIN), a frequent precursor to invasive cancer. Here, we have evaluated GLO1 expression by immunohistochemistry in archival tumor samples from 187 PCa patients (stage 2 and 3). Immunohistochemical analysis revealed GLO1 upregulation during tumor progression, observable in HGPIN and PCa versus normal prostatic tissue. GLO1 upregulation was identified as a novel hallmark of HGPIN lesions, displaying the highest staining intensity in all clinical patient specimens. GLO1 expression correlated with intermediate-high risk Gleason grade but not with patient age, biochemical recurrence, or pathological stage. Our data identify upregulated GLO1 expression as a molecular hallmark of HGPIN lesions detectable by immunohistochemical analysis. Since current pathological assessment of HGPIN status solely depends on morphological features, GLO1 may serve as a novel diagnostic marker that identifies this precancerous lesion.
Collapse
Affiliation(s)
- Liliana Rounds
- Department of Pharmacology and Toxicology, College of Pharmacy & UA Cancer Center, University of Arizona, Tucson, AZ 85724, USA; (L.R.); (J.J.)
- Roche Diagnostics Solutions, Tucson, AZ 85755, USA; (A.M.); (S.G.); (E.V.)
| | - Ray B. Nagle
- Department of Pathology, University of Arizona, Tucson, AZ 85724, USA;
| | - Andrea Muranyi
- Roche Diagnostics Solutions, Tucson, AZ 85755, USA; (A.M.); (S.G.); (E.V.)
| | - Jana Jandova
- Department of Pharmacology and Toxicology, College of Pharmacy & UA Cancer Center, University of Arizona, Tucson, AZ 85724, USA; (L.R.); (J.J.)
| | - Scott Gill
- Roche Diagnostics Solutions, Tucson, AZ 85755, USA; (A.M.); (S.G.); (E.V.)
| | - Elizabeth Vela
- Roche Diagnostics Solutions, Tucson, AZ 85755, USA; (A.M.); (S.G.); (E.V.)
| | - Georg T. Wondrak
- Department of Pharmacology and Toxicology, College of Pharmacy & UA Cancer Center, University of Arizona, Tucson, AZ 85724, USA; (L.R.); (J.J.)
| |
Collapse
|
26
|
Immunohistochemistry for Prostate Biopsy-Impact on Histological Prostate Cancer Diagnoses and Clinical Decision Making. ACTA ACUST UNITED AC 2021; 28:2123-2133. [PMID: 34207594 PMCID: PMC8293248 DOI: 10.3390/curroncol28030197] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/15/2021] [Accepted: 06/07/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND To test the value of immunohistochemistry (IHC) staining in prostate biopsies for changes in biopsy results and its impact on treatment decision-making. METHODS Between January 2017-June 2020, all patients undergoing prostate biopsies were identified and evaluated regarding additional IHC staining for diagnostic purpose. Final pathologic results after radical prostatectomy (RP) were analyzed regarding the effect of IHC at biopsy. RESULTS Of 606 biopsies, 350 (58.7%) received additional IHC staining. Of those, prostate cancer (PCa) was found in 208 patients (59.4%); while in 142 patients (40.6%), PCa could be ruled out through IHC. IHC patients harbored significantly more often Gleason 6 in biopsy (p < 0.01) and less suspicious baseline characteristics than patients without IHC. Of 185 patients with positive IHC and PCa detection, IHC led to a change in biopsy results in 81 (43.8%) patients. Of these patients with changes in biopsy results due to IHC, 42 (51.9%) underwent RP with 59.5% harboring ≥pT3 and/or Gleason 7-10. CONCLUSIONS Patients with IHC stains had less suspicious characteristics than patients without IHC. Moreover, in patients with positive IHC and PCa detection, a change in biopsy results was observed in >40%. Patients with changes in biopsy results partly underwent RP, in which 60% harbored significant PCa.
Collapse
|
27
|
Epstein JI, Amin MB, Fine SW, Algaba F, Aron M, Baydar DE, Beltran AL, Brimo F, Cheville JC, Colecchia M, Comperat E, da Cunha IW, Delprado W, DeMarzo AM, Giannico GA, Gordetsky JB, Guo CC, Hansel DE, Hirsch MS, Huang J, Humphrey PA, Jimenez RE, Khani F, Kong Q, Kryvenko ON, Kunju LP, Lal P, Latour M, Lotan T, Maclean F, Magi-Galluzzi C, Mehra R, Menon S, Miyamoto H, Montironi R, Netto GJ, Nguyen JK, Osunkoya AO, Parwani A, Robinson BD, Rubin MA, Shah RB, So JS, Takahashi H, Tavora F, Tretiakova MS, True L, Wobker SE, Yang XJ, Zhou M, Zynger DL, Trpkov K. The 2019 Genitourinary Pathology Society (GUPS) White Paper on Contemporary Grading of Prostate Cancer. Arch Pathol Lab Med 2021; 145:461-493. [PMID: 32589068 DOI: 10.5858/arpa.2020-0015-ra] [Citation(s) in RCA: 158] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2020] [Indexed: 11/06/2022]
Abstract
CONTEXT.— Controversies and uncertainty persist in prostate cancer grading. OBJECTIVE.— To update grading recommendations. DATA SOURCES.— Critical review of the literature along with pathology and clinician surveys. CONCLUSIONS.— Percent Gleason pattern 4 (%GP4) is as follows: (1) report %GP4 in needle biopsy with Grade Groups (GrGp) 2 and 3, and in needle biopsy on other parts (jars) of lower grade in cases with at least 1 part showing Gleason score (GS) 4 + 4 = 8; and (2) report %GP4: less than 5% or less than 10% and 10% increments thereafter. Tertiary grade patterns are as follows: (1) replace "tertiary grade pattern" in radical prostatectomy (RP) with "minor tertiary pattern 5 (TP5)," and only use in RP with GrGp 2 or 3 with less than 5% Gleason pattern 5; and (2) minor TP5 is noted along with the GS, with the GrGp based on the GS. Global score and magnetic resonance imaging (MRI)-targeted biopsies are as follows: (1) when multiple undesignated cores are taken from a single MRI-targeted lesion, an overall grade for that lesion is given as if all the involved cores were one long core; and (2) if providing a global score, when different scores are found in the standard and the MRI-targeted biopsy, give a single global score (factoring both the systematic standard and the MRI-targeted positive cores). Grade Groups are as follows: (1) Grade Groups (GrGp) is the terminology adopted by major world organizations; and (2) retain GS 3 + 5 = 8 in GrGp 4. Cribriform carcinoma is as follows: (1) report the presence or absence of cribriform glands in biopsy and RP with Gleason pattern 4 carcinoma. Intraductal carcinoma (IDC-P) is as follows: (1) report IDC-P in biopsy and RP; (2) use criteria based on dense cribriform glands (>50% of the gland is composed of epithelium relative to luminal spaces) and/or solid nests and/or marked pleomorphism/necrosis; (3) it is not necessary to perform basal cell immunostains on biopsy and RP to identify IDC-P if the results would not change the overall (highest) GS/GrGp part per case; (4) do not include IDC-P in determining the final GS/GrGp on biopsy and/or RP; and (5) "atypical intraductal proliferation (AIP)" is preferred for an intraductal proliferation of prostatic secretory cells which shows a greater degree of architectural complexity and/or cytological atypia than typical high-grade prostatic intraepithelial neoplasia, yet falling short of the strict diagnostic threshold for IDC-P. Molecular testing is as follows: (1) Ki67 is not ready for routine clinical use; (2) additional studies of active surveillance cohorts are needed to establish the utility of PTEN in this setting; and (3) dedicated studies of RNA-based assays in active surveillance populations are needed to substantiate the utility of these expensive tests in this setting. Artificial intelligence and novel grading schema are as follows: (1) incorporating reactive stromal grade, percent GP4, minor tertiary GP5, and cribriform/intraductal carcinoma are not ready for adoption in current practice.
Collapse
Affiliation(s)
- Jonathan I Epstein
- From the Departments of Pathology (Epstein, DeMarzo, Lotan), McGill University Health Center, Montréal, Quebec, Canada.,Urology (Epstein), David Geffen School of Medicine at UCLA, Los Angeles, California (Huang).,and Oncology (Epstein), The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Mahul B Amin
- Department of Pathology and Laboratory Medicine and Urology, University of Tennessee Health Science, Memphis (Amin)
| | - Samson W Fine
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (Fine)
| | - Ferran Algaba
- Department of Pathology, Fundacio Puigvert, Barcelona, Spain (Algaba)
| | - Manju Aron
- Department of Pathology, University of Southern California, Los Angeles (Aron)
| | - Dilek E Baydar
- Department of Pathology, Faculty of Medicine, Koç University, İstanbul, Turkey (Baydar)
| | - Antonio Lopez Beltran
- Department of Pathology, Champalimaud Centre for the Unknown, Lisbon, Portugal (Beltran)
| | - Fadi Brimo
- Department of Pathology, McGill University Health Center, Montréal, Quebec, Canada (Brimo)
| | - John C Cheville
- Department of Pathology, Mayo Clinic, Rochester, Minnesota (Cheville, Jimenez)
| | - Maurizio Colecchia
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy (Colecchia)
| | - Eva Comperat
- Department of Pathology, Hôpital Tenon, Sorbonne University, Paris, France (Comperat)
| | | | | | - Angelo M DeMarzo
- From the Departments of Pathology (Epstein, DeMarzo, Lotan), McGill University Health Center, Montréal, Quebec, Canada
| | - Giovanna A Giannico
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Giannico, Gordetsky)
| | - Jennifer B Gordetsky
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Giannico, Gordetsky)
| | - Charles C Guo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston (Guo)
| | - Donna E Hansel
- Department of Pathology, Oregon Health and Science University, Portland (Hansel)
| | - Michelle S Hirsch
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts (Hirsch)
| | - Jiaoti Huang
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California (Huang)
| | - Peter A Humphrey
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut (Humphrey)
| | - Rafael E Jimenez
- Department of Pathology, Mayo Clinic, Rochester, Minnesota (Cheville, Jimenez)
| | - Francesca Khani
- Department of Pathology and Laboratory Medicine and Urology, Weill Cornell Medicine, New York, New York (Khani, Robinson)
| | - Qingnuan Kong
- Department of Pathology, Qingdao Municipal Hospital, Qingdao, Shandong, China (Kong).,Kong is currently located at Kaiser Permanente Sacramento Medical Center, Sacramento, California
| | - Oleksandr N Kryvenko
- Departments of Pathology and Laboratory Medicine and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida (Kryvenko)
| | - L Priya Kunju
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan (Kunju, Mehra)
| | - Priti Lal
- Perelman School of Medicine, University of Pennsylvania, Philadelphia (Lal)
| | - Mathieu Latour
- Department of Pathology, CHUM, Université de Montréal, Montréal, Quebec, Canada (Latour)
| | - Tamara Lotan
- From the Departments of Pathology (Epstein, DeMarzo, Lotan), McGill University Health Center, Montréal, Quebec, Canada
| | - Fiona Maclean
- Douglass Hanly Moir Pathology, Faculty of Medicine and Health Sciences Macquarie University, North Ryde, Australia (Maclean)
| | - Cristina Magi-Galluzzi
- Department of Pathology, The University of Alabama at Birmingham, Birmingham (Magi-Galluzzi, Netto)
| | - Rohit Mehra
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan (Kunju, Mehra)
| | - Santosh Menon
- Department of Surgical Pathology, Tata Memorial Hospital, Parel, Mumbai, India (Menon)
| | - Hiroshi Miyamoto
- Departments of Pathology and Laboratory Medicine and Urology, University of Rochester Medical Center, Rochester, New York (Miyamoto)
| | - Rodolfo Montironi
- Section of Pathological Anatomy, School of Medicine, Polytechnic University of the Marche Region, United Hospitals, Ancona, Italy (Montironi)
| | - George J Netto
- Department of Pathology, The University of Alabama at Birmingham, Birmingham (Magi-Galluzzi, Netto)
| | - Jane K Nguyen
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio (Nguyen)
| | - Adeboye O Osunkoya
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia (Osunkoya)
| | - Anil Parwani
- Department of Pathology, Ohio State University, Columbus (Parwani, Zynger)
| | - Brian D Robinson
- Department of Pathology and Laboratory Medicine and Urology, Weill Cornell Medicine, New York, New York (Khani, Robinson)
| | - Mark A Rubin
- Department for BioMedical Research, University of Bern, Bern, Switzerland (Rubin)
| | - Rajal B Shah
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas (Shah)
| | - Jeffrey S So
- Institute of Pathology, St Luke's Medical Center, Quezon City and Global City, Philippines (So)
| | - Hiroyuki Takahashi
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan (Takahashi)
| | - Fabio Tavora
- Argos Laboratory, Federal University of Ceara, Fortaleza, Brazil (Tavora)
| | - Maria S Tretiakova
- Department of Pathology, University of Washington School of Medicine, Seattle (Tretiakova, True)
| | - Lawrence True
- Department of Pathology, University of Washington School of Medicine, Seattle (Tretiakova, True)
| | - Sara E Wobker
- Departments of Pathology and Laboratory Medicine and Urology, University of North Carolina, Chapel Hill (Wobker)
| | - Ximing J Yang
- Department of Pathology, Northwestern University, Chicago, Illinois (Yang)
| | - Ming Zhou
- Department of Pathology, Tufts Medical Center, Boston, Massachusetts (Zhou)
| | - Debra L Zynger
- Department of Pathology, Ohio State University, Columbus (Parwani, Zynger)
| | - Kiril Trpkov
- and Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Alberta, Canada (Trpkov)
| |
Collapse
|
28
|
Report From the International Society of Urological Pathology (ISUP) Consultation Conference on Molecular Pathology of Urogenital Cancers. I. Molecular Biomarkers in Prostate Cancer. Am J Surg Pathol 2020; 44:e15-e29. [PMID: 32044806 DOI: 10.1097/pas.0000000000001450] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The combined clinical and molecular heterogeneity of prostate cancer necessitates the use of prognostic, predictive, and diagnostic biomarkers to assist the clinician with treatment selection. The pathologist plays a critical role in guiding molecular biomarker testing in prostate cancer and requires a thorough knowledge of the current testing options. In the setting of clinically localized prostate cancer, prognostic biomarkers such as Ki-67 labeling, PTEN loss or mRNA-based genomic signatures can be useful to help determine whether definitive therapy is required. In the setting of advanced disease, predictive biomarkers, such as the presence of DNA repair deficiency mediated by BRCA2 loss or mismatch repair gene defects, may suggest the utility of poly-ADP ribosylase inhibition or immune checkpoint blockade. Finally, androgen receptor-related biomarkers or diagnostic biomarkers indicating the presence of small cell neuroendocrine prostate cancer may help guide the use of androgen receptor signaling inhibitors and chemotherapy. In this review, we examine the current evidence for several prognostic, predictive and diagnostic tissue-based molecular biomarkers in prostate cancer management. For each assay, we summarize a recent survey of the International Society of Urology Pathology (ISUP) members on current testing practices and include recommendations for testing that emerged from the ISUP Working Group on Molecular Pathology of Prostate Cancer and the 2019 Consultation Conference on Molecular Pathology of Urogenital Cancers.
Collapse
|
29
|
Lam D, Clark S, Stirzaker C, Pidsley R. Advances in Prognostic Methylation Biomarkers for Prostate Cancer. Cancers (Basel) 2020; 12:E2993. [PMID: 33076494 PMCID: PMC7602626 DOI: 10.3390/cancers12102993] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 12/24/2022] Open
Abstract
There is a major clinical need for accurate biomarkers for prostate cancer prognosis, to better inform treatment strategies and disease monitoring. Current clinically recognised prognostic factors, including prostate-specific antigen (PSA) levels, lack sensitivity and specificity in distinguishing aggressive from indolent disease, particularly in patients with localised intermediate grade prostate cancer. There has therefore been a major focus on identifying molecular biomarkers that can add prognostic value to existing markers, including investigation of DNA methylation, which has a known role in tumorigenesis. In this review, we will provide a comprehensive overview of the current state of DNA methylation biomarker studies in prostate cancer prognosis, and highlight the advances that have been made in this field. We cover the numerous studies into well-established candidate genes, and explore the technological transition that has enabled hypothesis-free genome-wide studies and the subsequent discovery of novel prognostic genes.
Collapse
Affiliation(s)
- Dilys Lam
- Epigenetics Research Laboratory, Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia; (D.L.); (S.C.); (C.S.)
| | - Susan Clark
- Epigenetics Research Laboratory, Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia; (D.L.); (S.C.); (C.S.)
- St. Vincent’s Clinical School, University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Clare Stirzaker
- Epigenetics Research Laboratory, Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia; (D.L.); (S.C.); (C.S.)
- St. Vincent’s Clinical School, University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Ruth Pidsley
- Epigenetics Research Laboratory, Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia; (D.L.); (S.C.); (C.S.)
- St. Vincent’s Clinical School, University of New South Wales, Sydney, New South Wales 2010, Australia
| |
Collapse
|
30
|
Darrell CM, Montironi R, Paner GP. Potential biomarkers and risk assessment models to enhance the tumor-node-metastasis (TNM) staging classification of urologic cancers. Expert Rev Mol Diagn 2020; 20:921-932. [PMID: 32876523 DOI: 10.1080/14737159.2020.1816827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION The anatomic-based TNM classification is considered the benchmark in cancer staging and has been regularly updated since its inception. In the current era of precision medicine, the added intention for future TNM modifications is to heighten its impact in the more 'personalized' level of cancer care. In urologic cancers, this goal may be achieved by incorporating 'non-anatomic' factors into TNM, such as biomarkers (e.g. gene alterations, molecular subtypes, genomic classifiers) and risk assessment models (e.g. nomogram, look-up table), while maintaining the anatomic extent as the foundation of staging. These different prognosticators can be combined and integrated, may serve as substratifiers for T, N, or M categories, and perhaps, incorporated as elements in TNM stage groupings to enhance their prognostic capability in urologic cancers. AREAS COVERED This review highlights candidate biomarkers and risk assessment models that can be explored to potentially improve TNM prognostication of bladder, prostate, kidney, and testicular cancers. EXPERT OPINION Recent advances in molecular analysis have increased the understanding of the genomic, transcriptomic, and epigenetic features for biomarker use in prognostication of urologic cancers, which together with the available risk assessment models, may complement and overcome the limitations of the traditional TNM staging.
Collapse
Affiliation(s)
- Caitlin M Darrell
- Departments of Pathology, Section of Urology, University of Chicago , Chicago, IL, USA
| | - Rodolfo Montironi
- School of Medicine, Section of Pathological Anatomy, Polytechnic University of the Marche Region , Ancona, Italy
| | - Gladell P Paner
- Departments of Pathology, Section of Urology, University of Chicago , Chicago, IL, USA.,Departments of Surgery, Section of Urology, University of Chicago , Chicago, IL, USA
| |
Collapse
|
31
|
van Leenders GJLH, Verhoef EI, Hollemans E. Prostate cancer growth patterns beyond the Gleason score: entering a new era of comprehensive tumour grading. Histopathology 2020; 77:850-861. [PMID: 32683729 PMCID: PMC7756302 DOI: 10.1111/his.14214] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 12/18/2022]
Abstract
The Gleason grading system is one of the most important factors in clinical decision‐making for prostate cancer patients, and is entirely based on the classification of tumour growth patterns. In recent years it has become clear that some individual growth patterns themselves have independent prognostic value, and could be used for better personalised risk stratification. In this review we summarise recent literature on the clinicopathological value and molecular characteristics of individual prostate cancer growth patterns, and show how these, most particularly cribriform architecture, could alter treatment decisions for prostate cancer patients.
Collapse
Affiliation(s)
| | - Esther I Verhoef
- Department of Pathology, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| | - Eva Hollemans
- Department of Pathology, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| |
Collapse
|
32
|
Farberg AS, Hall MA, Douglas L, Covington KR, Kurley SJ, Cook RW, Dinehart SM. Integrating gene expression profiling into NCCN high-risk cutaneous squamous cell carcinoma management recommendations: impact on patient management. Curr Med Res Opin 2020; 36:1301-1307. [PMID: 32351136 DOI: 10.1080/03007995.2020.1763284] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Objective: To integrate gene expression profiling into the management of high-risk cutaneous squamous cell carcinoma (cSCC) within the National Comprehensive Cancer Network (NCCN) guidelines to improve risk-aligned management recommendations.Methods: A cohort of 300 NCCN-defined high-risk cSCC patients, along with the American Joint Committee on Cancer (AJCC) T stage, Brigham and Women's Hospital (BWH) T stage, and known patient outcomes were analyzed. Risk classifications using a validated 40-gene expression profile (40-GEP) test and T stage were applied to NCCN patient management guidelines. Risk-directed patient management recommendations within the NCCN guidelines framework were aligned based on risk for metastasis.Results: Of the 300 NCCN high-risk cSCC patients, 159 (53.0%) were 40-GEP Class 1 and AJCC T1-T2, and 173 (57.7%) were Class 1 and BWH T1-2a, indicating low risk for metastasis and, thereby, suggesting low management intensity. The 40-GEP integration suggested high intensity management for only 24 (8.0%) patients (all Class 2B), and moderate intensity management for the remainder of the cohort.Conclusions: The 40-GEP test can be integrated within existing NCCN guideline recommendations for managing cSCC patients to help refine risk-directed management decisions. Integration of the 40-GEP test would allow >50% of this NCCN-defined high-risk cohort to be managed with the lowest intensity recommendations within the broad NCCN guidelines. High intensity management was deemed risk-appropriate for a small subpopulation (8.0%). This study demonstrates that the 40-GEP test, in combination with T stage, has clinical utility to impact patient management decisions in NCCN high-risk cSCC for improving risk-aligned management within the NCCN guidelines framework.
Collapse
Affiliation(s)
- Aaron S Farberg
- Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Dermatology, Arkansas Dermatology Skin Cancer Center, Little Rock, AR, USA
| | - Mary A Hall
- Research and Development, Castle Biosciences, Inc, Friendswood, TX, USA
| | - Leah Douglas
- Dermatology, Baylor College of Medicine, Houston, TX, USA
| | - Kyle R Covington
- Research and Development, Castle Biosciences, Inc, Friendswood, TX, USA
| | - Sarah J Kurley
- Research and Development, Castle Biosciences, Inc, Friendswood, TX, USA
| | - Robert W Cook
- Research and Development, Castle Biosciences, Inc, Friendswood, TX, USA
| | - Scott M Dinehart
- Dermatology, Arkansas Dermatology Skin Cancer Center, Little Rock, AR, USA
| |
Collapse
|
33
|
Wysong A, Newman JG, Covington KR, Kurley SJ, Ibrahim SF, Farberg AS, Bar A, Cleaver NJ, Somani AK, Panther D, Brodland DG, Zitelli J, Toyohara J, Maher IA, Xia Y, Bibee K, Griego R, Rigel DS, Meldi Plasseraud K, Estrada S, Sholl LM, Johnson C, Cook RW, Schmults CD, Arron ST. Validation of a 40-gene expression profile test to predict metastatic risk in localized high-risk cutaneous squamous cell carcinoma. J Am Acad Dermatol 2020; 84:361-369. [PMID: 32344066 DOI: 10.1016/j.jaad.2020.04.088] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/22/2020] [Accepted: 04/15/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND Current staging systems for cutaneous squamous cell carcinoma (cSCC) have limited positive predictive value for identifying patients who will experience metastasis. OBJECTIVE To develop and validate a gene expression profile (GEP) test for predicting risk for metastasis in localized, high-risk cSCC with the goal of improving risk-directed patient management. METHODS Archival formalin-fixed paraffin-embedded primary cSCC tissue and clinicopathologic data (n = 586) were collected from 23 independent centers in a prospectively designed study. A GEP signature was developed using a discovery cohort (n = 202) and validated in a separate, nonoverlapping, independent cohort (n = 324). RESULTS A prognostic 40-GEP test was developed and validated, stratifying patients with high-risk cSCC into classes based on metastasis risk: class 1 (low risk), class 2A (high risk), and class 2B (highest risk). For the validation cohort, 3-year metastasis-free survival rates were 91.4%, 80.6%, and 44.0%, respectively. A positive predictive value of 60% was achieved for the highest-risk group (class 2B), an improvement over staging systems, and negative predictive value, sensitivity, and specificity were comparable to staging systems. LIMITATIONS Potential understaging of cases could affect metastasis rate accuracy. CONCLUSION The 40-GEP test is an independent predictor of metastatic risk that can complement current staging systems for patients with high-risk cSCC.
Collapse
Affiliation(s)
- Ashley Wysong
- University of Nebraska Medical Center, Omaha, Nebraska
| | | | | | | | | | - Aaron S Farberg
- Icahn School of Medicine at Mount Sinai, New York, New York; Arkansas Dermatology Skin Cancer Center, Little Rock, Arkansas
| | - Anna Bar
- Oregon Health & Science University, Portland, Oregon
| | | | | | - David Panther
- Zitelli and Brodland, P.C. Skin Cancer Center, Pittsburgh, Pennsylvania
| | - David G Brodland
- Zitelli and Brodland, P.C. Skin Cancer Center, Pittsburgh, Pennsylvania
| | - John Zitelli
- Zitelli and Brodland, P.C. Skin Cancer Center, Pittsburgh, Pennsylvania
| | | | - Ian A Maher
- University of Minnesota, Minneapolis, Minnesota
| | - Yang Xia
- Brooke Army Medical Center, San Antonio, Texas
| | - Kristin Bibee
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | | | | | | | - Sarah Estrada
- Castle Biosciences, Inc, Phoenix, Arizona; Affiliated Dermatology, Scottsdale, Arizona
| | | | | | | | | | - Sarah T Arron
- University of California San Francisco, San Francisco, California.
| |
Collapse
|
34
|
Abstract
Artificial intelligence (AI) - the ability of a machine to perform cognitive tasks to achieve a particular goal based on provided data - is revolutionizing and reshaping our health-care systems. The current availability of ever-increasing computational power, highly developed pattern recognition algorithms and advanced image processing software working at very high speeds has led to the emergence of computer-based systems that are trained to perform complex tasks in bioinformatics, medical imaging and medical robotics. Accessibility to 'big data' enables the 'cognitive' computer to scan billions of bits of unstructured information, extract the relevant information and recognize complex patterns with increasing confidence. Computer-based decision-support systems based on machine learning (ML) have the potential to revolutionize medicine by performing complex tasks that are currently assigned to specialists to improve diagnostic accuracy, increase efficiency of throughputs, improve clinical workflow, decrease human resource costs and improve treatment choices. These characteristics could be especially helpful in the management of prostate cancer, with growing applications in diagnostic imaging, surgical interventions, skills training and assessment, digital pathology and genomics. Medicine must adapt to this changing world, and urologists, oncologists, radiologists and pathologists, as high-volume users of imaging and pathology, need to understand this burgeoning science and acknowledge that the development of highly accurate AI-based decision-support applications of ML will require collaboration between data scientists, computer researchers and engineers.
Collapse
|
35
|
Kremer A, Kremer T, Kristiansen G, Tolkach Y. Where is the limit of prostate cancer biomarker research? Systematic investigation of potential prognostic and diagnostic biomarkers. BMC Urol 2019; 19:46. [PMID: 31170942 PMCID: PMC6554887 DOI: 10.1186/s12894-019-0479-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 05/27/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The identification of appropriate biomarkers is essential to support important clinical decisions in patients with prostate cancer. The aim of our study was a systematic bioinformatical analysis of the mRNA expression of all genes available for the prostate adenocarcinoma cohort of The Cancer Genome Atlas (TCGA), regarding their potential prognostic and diagnostic role. METHODS The study cohort comprises 499 patients (TCGA prostate cancer cohort). mRNA expression data were available for approx. 20,000 genes. The bioinformatical statistical pipeline addressed gene expression differences in tumor vs. benign prostate tissue (including gene set enrichment analysis, GSEA) in samples from tumors with different aggressivenesses (Gleason score), as well as prognostic values in multistep survival analyses. RESULTS Among all genes analyzed, 1754 were significantly downregulated and 1553 genes were significantly upregulated in tumor tissue. In GSEA, 16 of 30 top enriched biological processes were alterations of epigenetic regulation at different levels. Significant correlation with Gleason Score was evident for 8724 genes (range of Pearson r-values 0.09-0.43; all p < 0.05). In univariate Cox regression analyses, mRNA expression of 3571 genes showed statistically significant association with biochemical recurrence-free survival with a range of hazard ratios 0.3-3.8 (p-value 7.4e- 07 to 0.05). Among these, 571 genes were independently associated with biochemical recurrence in multivariate analysis. Access to the full database including results is provided as supplement. CONCLUSIONS In our systematic analysis we found a big number of genes of potential diagnostic and prognostic value, many of which have not been studied in prostate cancer to date. Due to the comprehensive nature of this analysis and free access to the results, this study represents a reference database for prostate cancer researchers which can be used as a powerful tool for validation purposes and planning of new studies.
Collapse
Affiliation(s)
- Anika Kremer
- Institute of Pathology, University Hospital of Bonn, Bonn, Germany
| | - Tobias Kremer
- Institute of Computer Science, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Glen Kristiansen
- Institute of Pathology, University Hospital of Bonn, Bonn, Germany
| | - Yuri Tolkach
- Institute of Pathology, University Hospital of Bonn, Bonn, Germany.
| |
Collapse
|
36
|
Guo T, Li L, Zhong Q, Rupp NJ, Charmpi K, Wong CE, Wagner U, Rueschoff JH, Jochum W, Fankhauser CD, Saba K, Poyet C, Wild PJ, Aebersold R, Beyer A. Multi-region proteome analysis quantifies spatial heterogeneity of prostate tissue biomarkers. Life Sci Alliance 2018; 1. [PMID: 30090875 PMCID: PMC6078179 DOI: 10.26508/lsa.201800042] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Application of pressure cycling technology and Sequential Windowed Acquisition of all THeoretical mass spectrometry allows quantifying the degree of intra-tumor heterogeneity of protein expression in prostate tumors. The data show that protein intra-tumor heterogeneity, if not characterized, may distort protein biomarker suitability in tumor tissues. It remains unclear to what extent tumor heterogeneity impacts on protein biomarker discovery. Here, we quantified proteome intra-tissue heterogeneity (ITH) based on a multi-region analysis of prostate tissues using pressure cycling technology and Sequential Windowed Acquisition of all THeoretical fragment ion mass spectrometry. We quantified 6,873 proteins and analyzed the ITH of 3,700 proteins. The level of ITH varied depending on proteins and tissue types. Benign tissues exhibited more complex ITH patterns than malignant tissues. Spatial variability of 10 prostate biomarkers was validated by immunohistochemistry in an independent cohort (n = 83) using tissue microarrays. Prostate-specific antigen was preferentially variable in benign prostatic hyperplasia, whereas growth/differentiation factor 15 substantially varied in prostate adenocarcinomas. Furthermore, we found that DNA repair pathways exhibited a high degree of variability in tumorous tissues, which may contribute to the genetic heterogeneity of tumors. This study conceptually adds a new perspective to protein biomarker discovery: it suggests that recent technological progress should be exploited to quantify and account for spatial proteome variation to complement biomarker identification and utilization.
Collapse
Affiliation(s)
- Tiannan Guo
- Department of Biology, Institute of Molecular Systems Biology, ETH, Zurich, Switzerland.,Westlake Institute for Advanced Study, Westlake University, Hangzhou, Zhejiang, China
| | - Li Li
- CECAD, University of Cologne, Cologne, Germany
| | - Qing Zhong
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland.,Cancer Data Science Group, ProCan, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia
| | - Niels J Rupp
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | | | - Christine E Wong
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Ulrich Wagner
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Jan H Rueschoff
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Wolfram Jochum
- Institute of Pathology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | | | - Karim Saba
- Department of Urology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Cedric Poyet
- Department of Urology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Peter J Wild
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland.,Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, ETH, Zurich, Switzerland.,Faculty of Science, University of Zurich, Zurich, Switzerland
| | - Andreas Beyer
- CECAD, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| |
Collapse
|