1
|
Gallus R, Rizzo D, Rossi G, Mureddu L, Galli J, Artuso A, Bussu F. p16 Expression in Laryngeal Squamous Cell Carcinoma: A Surrogate or Independent Prognostic Marker? Pathogens 2024; 13:100. [PMID: 38392838 PMCID: PMC10892421 DOI: 10.3390/pathogens13020100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 01/21/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) is a common malignancy that, despite scientific advancements, has not seen an improvement in its prognosis in the last decades. Few promising predictive markers have been found and none are relevant in clinical practice. p16ink4a, an oncosuppressor protein involved in cell cycle arrest, with a prognostic impact on other cancers, has been widely used in the head and neck region as a surrogate marker of HPV infection. Published papers and recent meta-analyses seem to minimize the biological role of HPV in the context of LSCC's cancerogenesis, and to disprove the reliability of p16ink4a as a surrogate prognostic marker in this context, while still highlighting its potential role as an independent predictor of survival. Unfortunately, the available literature, in particular during the last two decades, is often not focused on its potential role as an independent biomarker and few relevant data are found in papers mainly focused on HPV. The available data suggest that future research should focus specifically on p16ink4a, taking into account both its potential inactivation and overexpression, different patterns of staining, and immunohistochemistry cutoffs, and should focus not on its potential role as a surrogate marker but on its independent role as a predictor of survival.
Collapse
Affiliation(s)
- Roberto Gallus
- Otolaryngology, Mater Olbia Hospital, 07026 Olbia, Italy; (R.G.); (A.A.)
| | - Davide Rizzo
- U.O.C. Otorinolaringoiatria, Azienda Ospedaliero Universitaria di Sassari, Viale San Pietro, 43, 07100 Sassari, Italy; (D.R.); (F.B.)
- Otolaryngology Division, Department of Medicine, Surgery and Pharmacology, University of Sassari, Viale San Pietro, 43, 07100 Sassari, Italy
| | - Giorgia Rossi
- Unit of Otorhinolaryngology and Head-Neck Surgery, “A. Gemelli” Hospital Foundation IRCCS, 00168 Rome, Italy; (G.R.); (J.G.)
| | - Luca Mureddu
- U.O.C. Otorinolaringoiatria, Azienda Ospedaliero Universitaria di Sassari, Viale San Pietro, 43, 07100 Sassari, Italy; (D.R.); (F.B.)
| | - Jacopo Galli
- Unit of Otorhinolaryngology and Head-Neck Surgery, “A. Gemelli” Hospital Foundation IRCCS, 00168 Rome, Italy; (G.R.); (J.G.)
- Department of Head-Neck and Sensory Organs, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Alberto Artuso
- Otolaryngology, Mater Olbia Hospital, 07026 Olbia, Italy; (R.G.); (A.A.)
| | - Francesco Bussu
- U.O.C. Otorinolaringoiatria, Azienda Ospedaliero Universitaria di Sassari, Viale San Pietro, 43, 07100 Sassari, Italy; (D.R.); (F.B.)
- Otolaryngology Division, Department of Medicine, Surgery and Pharmacology, University of Sassari, Viale San Pietro, 43, 07100 Sassari, Italy
| |
Collapse
|
2
|
Wilkins A, Gusterson B, Tovey H, Griffin C, Stuttle C, Daley F, Corbishley CM, Dearnaley D, Hall E, Somaiah N. Multi-candidate immunohistochemical markers to assess radiation response and prognosis in prostate cancer: results from the CHHiP trial of radiotherapy fractionation. EBioMedicine 2023; 88:104436. [PMID: 36708693 PMCID: PMC9900483 DOI: 10.1016/j.ebiom.2023.104436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/20/2022] [Accepted: 12/25/2022] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Protein markers of cellular proliferation, hypoxia, apoptosis, cell cycle checkpoints, growth factor signalling and inflammation in localised prostate tumours have previously shown prognostic ability. A translational substudy within the CHHiP trial of radiotherapy fractionation evaluated whether these could improve prediction of prognosis and assist treatment stratification following either conventional or hypofractionated radiotherapy. METHODS Using case:control methodology, patients with biochemical or clinical failure after radiotherapy (BCR) were matched to patients without recurrence according to established prognostic factors (Gleason score, presenting PSA, tumour-stage) and fractionation schedule. Immunohistochemical (IHC) staining of diagnostic biopsy sections was performed and scored for HIF1α, Bcl-2, Ki67, Geminin, p16, p53, p-chk1 and PTEN. Univariable and multivariable conditional logistic regression models, adjusted for matching strata and age, estimated the prognostic value of each IHC biomarker, including interaction terms to determine BCR prediction according to fractionation. FINDINGS IHC results were available for up to 336 tumours. PTEN, Geminin, mean Ki67 and max Ki67 were prognostic after adjusting for multiple comparisons and were fitted in a multivariable model (n = 212, 106 matched pairs). Here, PTEN and Geminin showed significant prediction of prognosis. No marker predicted BCR according to fractionation. INTERPRETATION Geminin or Ki67, and PTEN, predicted response to radiotherapy independently of established prognostic factors. These results provide essential independent external validation of previous findings and confirm a role for these markers in treatment stratification. FUNDING Cancer Research UK (BIDD) grant (A12518), Cancer Research UK (C8262/A7253), Department of Health, Prostate Cancer UK, Movember Foundation, NIHR Biomedical Research Centre at Royal Marsden/ICR.
Collapse
Affiliation(s)
- Anna Wilkins
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom; Royal Marsden Hospital, Sutton, United Kingdom.
| | - Barry Gusterson
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Holly Tovey
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Clare Griffin
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Christine Stuttle
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Frances Daley
- Division of Breast Cancer Research, The Institute of Cancer Research, London, United Kingdom
| | - Catherine M Corbishley
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - David Dearnaley
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom; Royal Marsden Hospital, Sutton, United Kingdom
| | - Emma Hall
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Navita Somaiah
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom; Royal Marsden Hospital, Sutton, United Kingdom
| |
Collapse
|
3
|
Wong M, Bierman Y, Pettaway C, Kittles R, Mims M, Jones J, Ittmann M. Comparative analysis of p16 expression among African American and European American prostate cancer patients. Prostate 2019; 79:1274-1283. [PMID: 31111520 PMCID: PMC6617792 DOI: 10.1002/pros.23833] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 05/01/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND Expression of p16 is increased in a number of malignancies, including prostate cancer (PCa). Recent studies in a European cohort showed that expression of p16 is correlated with expression of the TMPRSS2/ERG (T/E) fusion protein. The T/E fusion is significantly less common in PCas in African American (AA) men. Thus, it would be predicted that p16 expression should be less common in PCas in AA men. We, therefore, sought to compare the expression of p16 in benign prostate and PCas from AA and European American (EA) men. METHODS Immunohistochemistry for p16 and ERG was performed on tissue microarrays constructed from radical prostatectomies performed on AA and EA veterans. Staining was scored and the scores compared with demographic, clinical and pathological parameters. Percent of West African ancestry in the AA cohort was assessed using ancestry informative markers. RESULTS Contrary to our predictions, p16 expression was similar in the cancers in the AA and EA cohorts. Consistent with prior reports, expression of p16 was quite low in benign prostate tissues from EA patients but surprisingly was significantly higher in benign tissues from AA patients. Expression of p16 was significantly associated with a family history of PCa in AA men. In addition, p16 was associated with ERG expression in AA PCa. CONCLUSIONS While overall expression of p16 is similar in PCas from the two racial groups, the expression of p16 in benign tissues from a subset of AA men and the stronger correlation with ERG expression implies that there are different mechanisms for p16 overexpression in PCas from the two racial groups.
Collapse
Affiliation(s)
- Myra Wong
- Department of Pathology and Immunology, Michael E. DeBakey VA Medical CenterBaylor College of MedicineHoustonTexas
| | - Yaeli Bierman
- Department of Pathology and Immunology, Michael E. DeBakey VA Medical CenterBaylor College of MedicineHoustonTexas
| | - Curtis Pettaway
- Department of UrologyUT MD Anderson Cancer CenterHoustonTexas
| | - Rick Kittles
- Department of Population Sciences, Division of Health EquitiesCity of Hope Comprehensive Cancer CenterDuarteCalifornia
| | - Martha Mims
- Department of MedicineBaylor College of MedicineHoustonTexas
| | - Jeffrey Jones
- Scott Department of Urology, Michael E. DeBakey VA Medical CenterBaylor College of MedicineHoustonTexas
| | - Michael Ittmann
- Department of Pathology and Immunology, Michael E. DeBakey VA Medical CenterBaylor College of MedicineHoustonTexas
| |
Collapse
|
4
|
Lee DH, Yu EJ, Aldahl J, Yang J, He Y, Hooker E, Le V, Mi J, Olson A, Wu H, Geradts J, Xiao GQ, Gonzalgo ML, Cardiff RD, Sun Z. Deletion of the p16INK4a tumor suppressor and expression of the androgen receptor induce sarcomatoid carcinomas with signet ring cells in the mouse prostate. PLoS One 2019; 14:e0211153. [PMID: 30677079 PMCID: PMC6345450 DOI: 10.1371/journal.pone.0211153] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 01/08/2019] [Indexed: 12/26/2022] Open
Abstract
The tumor suppressor p16Ink4a, encoded by the INK4a gene, is an inhibitor of cyclin D-dependent kinases 4 and 6, CDK4 and CDK6. This inhibition prevents the phosphorylation of the retinoblastoma protein (pRb), resulting in cellular senescence through inhibition of E2F-mediated transcription of S phase genes required for cell proliferation. The p16Ink4a plays an important role in tumor suppression, whereby its deletion, mutation, or epigenetic silencing is a frequently observed genetic alteration in prostate cancer. To assess its roles and related molecular mechanisms in prostate cancer initiation and progression, we generated a mouse model with conditional deletion of p16Ink4a in prostatic luminal epithelium. The mice underwent oncogenic transformation and developed prostatic intraepithelial neoplasia (PIN) from eight months of age, but failed to develop prostatic tumors. Given the prevalence of aberrant androgen signaling pathways in prostate cancer initiation and progression, we then generated R26hARL/wt:p16L/L: PB-Cre4 compound mice, in which conditional expression of the human AR transgene and deletion of p16Ink4a co-occur in prostatic luminal epithelial cells. While R26hARL/wt:PB-Cre4 mice showed no visible pathological changes, R26hARL/wt:p16L/L: PB-Cre4 compound mice displayed an early onset of high-grade PIN (HGPIN), prostatic carcinoma, and metastatic lesions. Strikingly, we observed tumors resembling human sarcomatoid carcinoma with intermixed focal regions of signet ring cell carcinoma (SRCC) in the prostates of the compound mice. Further characterization of these tumors showed they were of luminal epithelial cell origin, and featured characteristics of epithelial to mesenchymal transition (EMT) with enhanced proliferative and invasive capabilities. Our results not only implicate a biological role for AR expression and p16Ink4a deletion in the pathogenesis of prostatic SRCC, but also provide a new and unique genetically engineered mouse (GEM) model for investigating the molecular mechanisms for SRCC development.
Collapse
Affiliation(s)
- Dong-Hong Lee
- Department of Cancer Biology, Beckman Research Institute, City of Hope, Duarte, California, United States of America
| | - Eun-Jeong Yu
- Department of Cancer Biology, Beckman Research Institute, City of Hope, Duarte, California, United States of America
| | - Joseph Aldahl
- Department of Cancer Biology, Beckman Research Institute, City of Hope, Duarte, California, United States of America
| | - Julie Yang
- Department of Cancer Biology, Beckman Research Institute, City of Hope, Duarte, California, United States of America
| | - Yongfeng He
- Department of Cancer Biology, Beckman Research Institute, City of Hope, Duarte, California, United States of America
| | - Erika Hooker
- Department of Cancer Biology, Beckman Research Institute, City of Hope, Duarte, California, United States of America
| | - Vien Le
- Department of Cancer Biology, Beckman Research Institute, City of Hope, Duarte, California, United States of America
| | - Jiaqi Mi
- Department of Cancer Biology, Beckman Research Institute, City of Hope, Duarte, California, United States of America
| | - Adam Olson
- Department of Cancer Biology, Beckman Research Institute, City of Hope, Duarte, California, United States of America
| | - Huiqing Wu
- Department of Pathology, Beckman Research Institute, City of Hope, Duarte, California, United States of America
| | - Joseph Geradts
- Department of Population Sciences, Beckman Research Institute, City of Hope, Duarte, California, United States of America
| | - Guang Q. Xiao
- Department of Pathology, Keck Medical School, University of South California, Los Angeles, California, United States of America
| | - Mark L. Gonzalgo
- Department of Urology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Robert D. Cardiff
- Comparative Medicine, University of California at Davis, Davis, California, United States of America
| | - Zijie Sun
- Department of Cancer Biology, Beckman Research Institute, City of Hope, Duarte, California, United States of America
- * E-mail:
| |
Collapse
|
5
|
Rambau PF, Vierkant RA, Intermaggio MP, Kelemen LE, Goodman MT, Herpel E, Pharoah PD, Kommoss S, Jimenez‐Linan M, Karlan BY, Gentry‐Maharaj A, Menon U, Polo SH, Candido dos Reis FJ, Doherty JA, Gayther SA, Sharma R, Larson MC, Harnett PR, Hatfield E, de Andrade JM, Nelson GS, Steed H, Schildkraut JM, Carney ME, Høgdall E, Whittemore AS, Widschwendter M, Kennedy CJ, Wang F, Wang Q, Wang C, Armasu SM, Daley F, Coulson P, Jones ME, Anglesio MS, Chow C, de Fazio A, García‐Closas M, Brucker SY, Cybulski C, Harris HR, Hartkopf AD, Huzarski T, Jensen A, Lubiński J, Oszurek O, Benitez J, Mina F, Staebler A, Taran FA, Pasternak J, Talhouk A, Rossing MA, Hendley J, AOCS Group, Edwards RP, Fereday S, Modugno F, Ness RB, Sieh W, El‐Bahrawy MA, Winham SJ, Lester J, Kjaer SK, Gronwald J, Sinn P, Fasching PA, Chang‐Claude J, Moysich KB, Bowtell DD, Hernandez BY, Luk H, Behrens S, Shah M, Jung A, Ghatage P, Alsop J, Alsop K, García‐Donas J, Thompson PJ, Swerdlow AJ, Karpinskyj C, Cazorla‐Jiménez A, García MJ, Deen S, Wilkens LR, Palacios J, Berchuck A, Koziak JM, Brenton JD, Cook LS, Goode EL, Huntsman DG, Ramus SJ, Köbel M. Association of p16 expression with prognosis varies across ovarian carcinoma histotypes: an Ovarian Tumor Tissue Analysis consortium study. J Pathol Clin Res 2018; 4:250-261. [PMID: 30062862 PMCID: PMC6174617 DOI: 10.1002/cjp2.109] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 06/29/2018] [Accepted: 07/16/2018] [Indexed: 12/24/2022]
Abstract
We aimed to validate the prognostic association of p16 expression in ovarian high-grade serous carcinomas (HGSC) and to explore it in other ovarian carcinoma histotypes. p16 protein expression was assessed by clinical-grade immunohistochemistry in 6525 ovarian carcinomas including 4334 HGSC using tissue microarrays from 24 studies participating in the Ovarian Tumor Tissue Analysis consortium. p16 expression patterns were interpreted as abnormal (either overexpression referred to as block expression or absence) or normal (heterogeneous). CDKN2A (which encodes p16) mRNA expression was also analyzed in a subset (n = 2280) mostly representing HGSC (n = 2010). Association of p16 expression with overall survival (OS) was determined within histotypes as was CDKN2A expression for HGSC only. p16 block expression was most frequent in HGSC (56%) but neither protein nor mRNA expression was associated with OS. However, relative to heterogeneous expression, block expression was associated with shorter OS in endometriosis-associated carcinomas, clear cell [hazard ratio (HR): 2.02, 95% confidence (CI) 1.47-2.77, p < 0.001] and endometrioid (HR: 1.88, 95% CI 1.30-2.75, p = 0.004), while absence was associated with shorter OS in low-grade serous carcinomas (HR: 2.95, 95% CI 1.61-5.38, p = 0.001). Absence was most frequent in mucinous carcinoma (50%), and was not associated with OS in this histotype. The prognostic value of p16 expression is histotype-specific and pattern dependent. We provide definitive evidence against an association of p16 expression with survival in ovarian HGSC as previously suggested. Block expression of p16 in clear cell and endometrioid carcinoma should be further validated as a prognostic marker, and absence in low-grade serous carcinoma justifies CDK4 inhibition.
Collapse
Affiliation(s)
- Peter F Rambau
- Department of Pathology and Laboratory MedicineUniversity of Calgary, Foothills Medical CenterCalgaryABCanada
- Pathology DepartmentCatholic University of Health and Allied Sciences‐BugandoMwanzaTanzania
| | - Robert A Vierkant
- Department of Health Sciences Research, Division of Biomedical Statistics and InformaticsMayo ClinicRochesterMNUSA
| | - Maria P Intermaggio
- School of Women's and Children's HealthFaculty of Medicine, University of NSW SydneySydneyNSWAustralia
| | - Linda E Kelemen
- Department of Public Health SciencesMedical University of South CarolinaCharlestonSCUSA
| | - Marc T Goodman
- Samuel Oschin Comprehensive Cancer Institute, Cancer Prevention and Genetics Program, Cedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Esther Herpel
- National Center for Tumor Diseases, University of HeidelbergHeidelbergGermany
| | - Paul D Pharoah
- Centre for Cancer Genetic Epidemiology, Department of OncologyUniversity of CambridgeCambridgeUK
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary CareUniversity of CambridgeCambridgeUK
| | - Stefan Kommoss
- Department of Women's HealthTübingen University HospitalTübingenGermany
| | | | - Beth Y Karlan
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Aleksandra Gentry‐Maharaj
- Gynaecological Cancer Research Centre, Women's Cancer, Institute for Women's Health, University College LondonLondonUK
| | - Usha Menon
- Gynaecological Cancer Research Centre, Women's Cancer, Institute for Women's Health, University College LondonLondonUK
| | | | - Francisco J Candido dos Reis
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical SchoolUniversity of São PauloRibeirão PretoBrazil
| | - Jennifer Anne Doherty
- Department of Population Health SciencesHuntsman Cancer Institute, University of UtahSalt Lake CityUTUSA
| | - Simon A Gayther
- Department of Preventive Medicine, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCAUSA
- Center for Cancer Prevention and Translational GenomicsSamuel Oschin Comprehensive Cancer Institute, Cedars‐Sinai Medical CenterLos AngelesCAUSA
- Department of Biomedical SciencesCedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Raghwa Sharma
- Pathology West ICPMR WestmeadWestmead Hospital, The University of SydneySydneyNSWAustralia
- University of Western Sydney at Westmead HospitalWestmeadNSWAustralia
| | - Melissa C Larson
- Department of Health Sciences Research, Division of Biomedical Statistics and InformaticsMayo ClinicRochesterMNUSA
| | - Paul R Harnett
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of SydneySydneyNSWAustralia
- The Crown Princess Mary Cancer Centre Westmead, Sydney‐West Cancer Network, Westmead HospitalSydneyNSWAustralia
| | - Emma Hatfield
- Department of Pathology and Laboratory MedicineUniversity of Calgary, Foothills Medical CenterCalgaryABCanada
| | - Jurandyr M de Andrade
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical SchoolUniversity of São PauloRibeirão PretoBrazil
| | - Gregg S Nelson
- Department of Oncology, Division of Gynecologic Oncology, Cumming School of MedicineUniversity of CalgaryCalgaryABCanada
| | - Helen Steed
- Department of Obstetrics and Gynecology, Division of Gynecologic OncologyRoyal Alexandra HospitalEdmontonABCanada
| | | | - Micheal E Carney
- John A. Burns School of Medicine, Department of Obstetrics and GynecologyUniversity of HawaiiHonoluluHIUSA
| | - Estrid Høgdall
- Department of Virus, Lifestyle and GenesDanish Cancer Society Research CenterCopenhagenDenmark
- Molecular Unit, Department of PathologyHerlev Hospital, University of CopenhagenCopenhagenDenmark
| | - Alice S Whittemore
- Department of Health Research and Policy – EpidemiologyStanford University School of MedicineStanfordCAUSA
- Department of Biomedical Data ScienceStanford University School of MedicineStanfordCAUSA
| | - Martin Widschwendter
- Gynaecological Cancer Research Centre, Women's Cancer, Institute for Women's Health, University College LondonLondonUK
| | - Catherine J Kennedy
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of SydneySydneyNSWAustralia
- Department of Gynaecological OncologyWestmead HospitalSydneyNSWAustralia
| | - Frances Wang
- Cancer Control and Population SciencesDuke Cancer InstituteDurhamNCUSA
- Department of Community and Family MedicineDuke University Medical CenterDurhamNCUSA
| | - Qin Wang
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary CareUniversity of CambridgeCambridgeUK
| | - Chen Wang
- Department of Health Sciences ResearchMayo ClinicRochesterMNUSA
| | - Sebastian M Armasu
- Department of Health Sciences Research, Division of Biomedical Statistics and InformaticsMayo ClinicRochesterMNUSA
| | - Frances Daley
- Division of Breast Cancer ResearchInstitute of Cancer ResearchLondonUK
- Division of BioscienceBrunel UniversityLondonUK
| | - Penny Coulson
- Division of Genetics and EpidemiologyInstitute of Cancer ResearchLondonUK
| | - Micheal E Jones
- Division of Genetics and EpidemiologyInstitute of Cancer ResearchLondonUK
| | - Micheal S Anglesio
- Department of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverBCCanada
| | - Christine Chow
- Genetic Pathology Evaluation Centre, Vancouver General Hospital and University of British ColumbiaVancouverBCCanada
| | - Anna de Fazio
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of SydneySydneyNSWAustralia
- Department of Gynaecological OncologyWestmead HospitalSydneyNSWAustralia
| | - Montserrat García‐Closas
- Division of Genetics and EpidemiologyInstitute of Cancer ResearchLondonUK
- Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMDUSA
| | - Sara Y Brucker
- Department of Gynecology and ObstetricsUniversity of TübingenTübingenGermany
| | - Cezary Cybulski
- Department of Genetics and PathologyPomeranian Medical UniversitySzczecinPoland
| | - Holly R Harris
- Program in Epidemiology, Division of Public Health SciencesFred Hutchinson Cancer Research CenterSeattleWAUSA
- Department of Environmental Medicine, Division of Nutritional EpidemiologyKarolinska InstitutetStockholmSweden
| | | | - Tomasz Huzarski
- Department of Genetics and PathologyPomeranian Medical UniversitySzczecinPoland
| | - Allan Jensen
- Department of Virus, Lifestyle and GenesDanish Cancer Society Research CenterCopenhagenDenmark
| | - Jan Lubiński
- Department of Genetics and PathologyPomeranian Medical UniversitySzczecinPoland
| | - Oleg Oszurek
- International Hereditary Cancer Center, Department of Genetics and PathologyPomeranian Medical UniversitySzczecinPoland
| | - Javier Benitez
- Human Cancer Genetics ProgrammeSpanish National Cancer Research Centre (CNIO)MadridSpain
- Biomedical Network on Rare Diseases (CIBERER)MadridSpain
| | - Fady Mina
- Department of Pathology and Laboratory MedicineUniversity of Calgary, Foothills Medical CenterCalgaryABCanada
| | - Annette Staebler
- Institute of Pathology, Tübingen University HospitalTübingenGermany
| | | | - Jana Pasternak
- Department of Women's HealthTübingen University HospitalTübingenGermany
| | - Aline Talhouk
- British Columbia's Ovarian Cancer Research (OVCARE) ProgramVancouver General Hospital, BC Cancer Agency and University of British ColumbiaVancouverBCCanada
| | - Mary Anne Rossing
- Program in Epidemiology, Division of Public Health SciencesFred Hutchinson Cancer Research CenterSeattleWAUSA
- Department of EpidemiologyUniversity of WashingtonSeattleWAUSA
| | - Joy Hendley
- Department of Research, Cancer Genomics and GeneticsPeter MacCallum Cancer CenterMelbourneVICAustralia
| | - AOCS Group
- Peter MacCallum Cancer CenterMelbourneVICAustralia
- Department of Genetics and Computational BiologyQIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia
| | - Robert P Edwards
- Ovarian Cancer Center of Excellence, Womens Cancer Research ProgramMagee‐Womens Research Institute and University of Pittsburgh Cancer InstitutePittsburghPAUSA
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive SciencesUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Sian Fereday
- Department of Research, Cancer Genomics and GeneticsPeter MacCallum Cancer CenterMelbourneVICAustralia
| | - Francesmary Modugno
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive SciencesUniversity of Pittsburgh School of MedicinePittsburghPAUSA
- Womens Cancer Research Center, Magee‐Womens Research Institute and Hillman Cancer CenterPittsburghPAUSA
| | - Roberta B Ness
- University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Weiva Sieh
- Department of Genetics and Genomic Sciences, Department of Population Health Science and Policy, Icahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Mona A El‐Bahrawy
- Department of Histopathology, Imperial College LondonHammersmith HospitalLondonUK
| | - Stacey J Winham
- Department of Health Sciences Research, Division of Biomedical Statistics and InformaticsMayo ClinicRochesterMNUSA
| | - Jenny Lester
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Susanne K Kjaer
- Department of Virus, Lifestyle and GenesDanish Cancer Society Research CenterCopenhagenDenmark
- Department of Gynaecology, RigshospitaletUniversity of CopenhagenCopenhagenDenmark
| | - Jacek Gronwald
- Department of Genetics and PathologyPomeranian Medical UniversitySzczecinPoland
| | - Peter Sinn
- Department of PathologyInstitute of Pathology, University Hospital HeidelbergHeidelbergGermany
| | - Peter A Fasching
- David Geffen School of Medicine, Department of Medicine Division of Hematology and OncologyUniversity of California at Los AngelesLos AngelesCAUSA
- Department of Gynecology and ObstetricsComprehensive Cancer Center ER‐EMN, University Hospital Erlangen, Friedrich‐Alexander‐University Erlangen‐NurembergErlangenGermany
| | - Jenny Chang‐Claude
- Division of Cancer EpidemiologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Cancer Epidemiology GroupUniversity Cancer Center Hamburg (UCCH), University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Kirsten B Moysich
- Division of Cancer Prevention and ControlRoswell Park Cancer InstituteBuffaloNYUSA
| | - David D Bowtell
- Department of Research, Cancer Genomics and GeneticsPeter MacCallum Cancer CenterMelbourneVICAustralia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneParkvilleVICAustralia
| | - Brenda Y Hernandez
- Cancer Epidemiology ProgramUniversity of Hawaii Cancer CenterHonoluluHIUSA
| | - Hugh Luk
- Cancer Epidemiology ProgramUniversity of Hawaii Cancer CenterHonoluluHIUSA
| | - Sabine Behrens
- Division of Cancer EpidemiologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Mitul Shah
- Centre for Cancer Genetic Epidemiology, Department of OncologyUniversity of CambridgeCambridgeUK
| | - Audrey Jung
- Division of Cancer EpidemiologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Prafull Ghatage
- Department of Oncology, Division of Gynecologic Oncology, Cumming School of MedicineUniversity of CalgaryCalgaryABCanada
| | - Jennifer Alsop
- Centre for Cancer Genetic Epidemiology, Department of OncologyUniversity of CambridgeCambridgeUK
| | - Kathryn Alsop
- Department of Research, Cancer Genomics and GeneticsPeter MacCallum Cancer CenterMelbourneVICAustralia
| | - Jesús García‐Donas
- Medical Oncology ServiceHM Hospitales – Centro Integral Oncológico HM Clara CampalMadridSpain
| | - Pamela J Thompson
- Samuel Oschin Comprehensive Cancer Institute, Cancer Prevention and Genetics Program, Cedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Anthony J Swerdlow
- Division of Genetics and EpidemiologyThe Institute of Cancer ResearchLondonUK
- Division of Breast Cancer ResearchThe Institute of Cancer ResearchLondonUK
| | - Chloe Karpinskyj
- Gynaecological Cancer Research Centre, Women's Cancer, Institute for Women's Health, University College LondonLondonUK
| | | | - María J García
- Human Cancer Genetics ProgrammeSpanish National Cancer Research Centre (CNIO)MadridSpain
- Biomedical Network on Rare Diseases (CIBERER)MadridSpain
| | - Susha Deen
- Department of HistopathologyQueen's Medical Centre, Nottingham University Hospitals NHS TrustNottinghamUK
| | - Lynne R Wilkens
- Cancer Epidemiology ProgramUniversity of Hawaii Cancer CenterHonoluluHIUSA
| | - José Palacios
- Pathology Department, IRYCIS, CIBERONCUniversidad de Alcalá, Hospital Universitario Ramón y CajalMadridSpain
| | - Andrew Berchuck
- Department of Obstetrics and GynecologyDuke University Medical CenterDurhamNCUSA
| | | | - James D Brenton
- Cancer Research UK Cambridge Institute, University of CambridgeCambridgeUK
| | - Linda S Cook
- University of New Mexico Health Sciences Center, University of New MexicoAlbuquerqueNMUSA
- Department of Cancer Epidemiology and Prevention ResearchAlberta Health ServicesCalgaryABCanada
| | - Ellen L Goode
- Department of Health Science Research, Division of EpidemiologyMayo ClinicRochesterMNUSA
| | - David G Huntsman
- Department of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverBCCanada
- British Columbia's Ovarian Cancer Research (OVCARE) ProgramVancouver General Hospital, BC Cancer Agency and University of British ColumbiaVancouverBCCanada
- Department of Molecular OncologyBC Cancer Agency Research CentreVancouverBCCanada
| | - Susan J Ramus
- School of Women's and Children's HealthFaculty of Medicine, University of NSW SydneySydneyNSWAustralia
- The Kinghorn Cancer Centre, Garvan Institute of Medical ResearchSydneyNSWAustralia
| | - Martin Köbel
- Department of Pathology and Laboratory MedicineUniversity of Calgary, Foothills Medical CenterCalgaryABCanada
| |
Collapse
|
6
|
Rambau PF, Vierkant RA, Intermaggio MP, Kelemen LE, Goodman MT, Herpel E, Pharoah PD, Kommoss S, Jimenez-Linan M, Karlan BY, Gentry-Maharaj A, Menon U, Polo SH, Candido Dos Reis FJ, Doherty JA, Gayther SA, Sharma R, Larson MC, Harnett PR, Hatfield E, de Andrade JM, Nelson GS, Steed H, Schildkraut JM, Carney ME, Høgdall E, Whittemore AS, Widschwendter M, Kennedy CJ, Wang F, Wang Q, Wang C, Armasu SM, Daley F, Coulson P, Jones ME, Anglesio MS, Chow C, de Fazio A, García-Closas M, Brucker SY, Cybulski C, Harris HR, Hartkopf AD, Huzarski T, Jensen A, Lubiński J, Oszurek O, Benitez J, Mina F, Staebler A, Taran FA, Pasternak J, Talhouk A, Rossing MA, Hendley J, Edwards RP, Fereday S, Modugno F, Ness RB, Sieh W, El-Bahrawy MA, Winham SJ, Lester J, Kjaer SK, Gronwald J, Sinn P, Fasching PA, Chang-Claude J, Moysich KB, Bowtell DD, Hernandez BY, Luk H, Behrens S, Shah M, Jung A, Ghatage P, Alsop J, Alsop K, García-Donas J, Thompson PJ, Swerdlow AJ, Karpinskyj C, Cazorla-Jiménez A, García MJ, Deen S, Wilkens LR, Palacios J, Berchuck A, Koziak JM, Brenton JD, Cook LS, Goode EL, Huntsman DG, Ramus SJ, Köbel M. Association of p16 expression with prognosis varies across ovarian carcinoma histotypes: an Ovarian Tumor Tissue Analysis consortium study. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2018. [PMID: 30062862 DOI: 10.1002/cjp2.109] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We aimed to validate the prognostic association of p16 expression in ovarian high-grade serous carcinomas (HGSC) and to explore it in other ovarian carcinoma histotypes. p16 protein expression was assessed by clinical-grade immunohistochemistry in 6525 ovarian carcinomas including 4334 HGSC using tissue microarrays from 24 studies participating in the Ovarian Tumor Tissue Analysis consortium. p16 expression patterns were interpreted as abnormal (either overexpression referred to as block expression or absence) or normal (heterogeneous). CDKN2A (which encodes p16) mRNA expression was also analyzed in a subset (n = 2280) mostly representing HGSC (n = 2010). Association of p16 expression with overall survival (OS) was determined within histotypes as was CDKN2A expression for HGSC only. p16 block expression was most frequent in HGSC (56%) but neither protein nor mRNA expression was associated with OS. However, relative to heterogeneous expression, block expression was associated with shorter OS in endometriosis-associated carcinomas, clear cell [hazard ratio (HR): 2.02, 95% confidence (CI) 1.47-2.77, p < 0.001] and endometrioid (HR: 1.88, 95% CI 1.30-2.75, p = 0.004), while absence was associated with shorter OS in low-grade serous carcinomas (HR: 2.95, 95% CI 1.61-5.38, p = 0.001). Absence was most frequent in mucinous carcinoma (50%), and was not associated with OS in this histotype. The prognostic value of p16 expression is histotype-specific and pattern dependent. We provide definitive evidence against an association of p16 expression with survival in ovarian HGSC as previously suggested. Block expression of p16 in clear cell and endometrioid carcinoma should be further validated as a prognostic marker, and absence in low-grade serous carcinoma justifies CDK4 inhibition.
Collapse
Affiliation(s)
- Peter F Rambau
- Department of Pathology and Laboratory Medicine, University of Calgary, Foothills Medical Center, Calgary, AB, Canada.,Pathology Department, Catholic University of Health and Allied Sciences-Bugando, Mwanza, Tanzania
| | - Robert A Vierkant
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Maria P Intermaggio
- School of Women's and Children's Health, Faculty of Medicine, University of NSW Sydney, Sydney, NSW, Australia
| | - Linda E Kelemen
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Marc T Goodman
- Samuel Oschin Comprehensive Cancer Institute, Cancer Prevention and Genetics Program, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Esther Herpel
- National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany
| | - Paul D Pharoah
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK.,Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Stefan Kommoss
- Department of Women's Health, Tübingen University Hospital, Tübingen, Germany
| | | | - Beth Y Karlan
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Aleksandra Gentry-Maharaj
- Gynaecological Cancer Research Centre, Women's Cancer, Institute for Women's Health, University College London, London, UK
| | - Usha Menon
- Gynaecological Cancer Research Centre, Women's Cancer, Institute for Women's Health, University College London, London, UK
| | - Susanna Hernando Polo
- Medical Oncology Service, Hospital Universitario Funcación Alcorcón, Alcorcón, Spain
| | - Francisco J Candido Dos Reis
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Jennifer Anne Doherty
- Department of Population Health Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Simon A Gayther
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Center for Cancer Prevention and Translational Genomics, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Raghwa Sharma
- Pathology West ICPMR Westmead, Westmead Hospital, The University of Sydney, Sydney, NSW, Australia.,University of Western Sydney at Westmead Hospital, Westmead, NSW, Australia
| | - Melissa C Larson
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Paul R Harnett
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia.,The Crown Princess Mary Cancer Centre Westmead, Sydney-West Cancer Network, Westmead Hospital, Sydney, NSW, Australia
| | - Emma Hatfield
- Department of Pathology and Laboratory Medicine, University of Calgary, Foothills Medical Center, Calgary, AB, Canada
| | - Jurandyr M de Andrade
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Gregg S Nelson
- Department of Oncology, Division of Gynecologic Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Helen Steed
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Royal Alexandra Hospital, Edmonton, AB, Canada
| | - Joellen M Schildkraut
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - Micheal E Carney
- John A. Burns School of Medicine, Department of Obstetrics and Gynecology, University of Hawaii, Honolulu, HI, USA
| | - Estrid Høgdall
- Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark.,Molecular Unit, Department of Pathology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Alice S Whittemore
- Department of Health Research and Policy - Epidemiology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Martin Widschwendter
- Gynaecological Cancer Research Centre, Women's Cancer, Institute for Women's Health, University College London, London, UK
| | - Catherine J Kennedy
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia.,Department of Gynaecological Oncology, Westmead Hospital, Sydney, NSW, Australia
| | - Frances Wang
- Cancer Control and Population Sciences, Duke Cancer Institute, Durham, NC, USA.,Department of Community and Family Medicine, Duke University Medical Center, Durham, NC, USA
| | - Qin Wang
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Chen Wang
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Sebastian M Armasu
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Frances Daley
- Division of Breast Cancer Research, Institute of Cancer Research, London, UK.,Division of Bioscience, Brunel University, London, UK
| | - Penny Coulson
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - Micheal E Jones
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - Micheal S Anglesio
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Christine Chow
- Genetic Pathology Evaluation Centre, Vancouver General Hospital and University of British Columbia, Vancouver, BC, Canada
| | - Anna de Fazio
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia.,Department of Gynaecological Oncology, Westmead Hospital, Sydney, NSW, Australia
| | - Montserrat García-Closas
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK.,Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Sara Y Brucker
- Department of Gynecology and Obstetrics, University of Tübingen, Tübingen, Germany
| | - Cezary Cybulski
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Holly R Harris
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Department of Environmental Medicine, Division of Nutritional Epidemiology, Karolinska Institutet, Stockholm, Sweden
| | - Andreas D Hartkopf
- Department of Women's Health, Tübingen University Hospital, Tübingen, Germany
| | - Tomasz Huzarski
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Allan Jensen
- Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Jan Lubiński
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Oleg Oszurek
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Javier Benitez
- Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.,Biomedical Network on Rare Diseases (CIBERER), Madrid, Spain
| | - Fady Mina
- Department of Pathology and Laboratory Medicine, University of Calgary, Foothills Medical Center, Calgary, AB, Canada
| | - Annette Staebler
- Institute of Pathology, Tübingen University Hospital, Tübingen, Germany
| | - Florin Andrei Taran
- Department of Women's Health, Tübingen University Hospital, Tübingen, Germany
| | - Jana Pasternak
- Department of Women's Health, Tübingen University Hospital, Tübingen, Germany
| | - Aline Talhouk
- British Columbia's Ovarian Cancer Research (OVCARE) Program, Vancouver General Hospital, BC Cancer Agency and University of British Columbia, Vancouver, BC, Canada
| | - Mary Anne Rossing
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Joy Hendley
- Department of Research, Cancer Genomics and Genetics, Peter MacCallum Cancer Center, Melbourne, VIC, Australia
| | -
- Peter MacCallum Cancer Center, Melbourne, VIC, Australia.,Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Robert P Edwards
- Ovarian Cancer Center of Excellence, Womens Cancer Research Program, Magee-Womens Research Institute and University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA.,Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sian Fereday
- Department of Research, Cancer Genomics and Genetics, Peter MacCallum Cancer Center, Melbourne, VIC, Australia
| | - Francesmary Modugno
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Womens Cancer Research Center, Magee-Womens Research Institute and Hillman Cancer Center, Pittsburgh, PA, USA
| | - Roberta B Ness
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Weiva Sieh
- Department of Genetics and Genomic Sciences, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mona A El-Bahrawy
- Department of Histopathology, Imperial College London, Hammersmith Hospital, London, UK
| | - Stacey J Winham
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Jenny Lester
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Susanne K Kjaer
- Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark.,Department of Gynaecology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jacek Gronwald
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Peter Sinn
- Department of Pathology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Peter A Fasching
- David Geffen School of Medicine, Department of Medicine Division of Hematology and Oncology, University of California at Los Angeles, Los Angeles, CA, USA.,Department of Gynecology and Obstetrics, Comprehensive Cancer Center ER-EMN, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Cancer Epidemiology Group, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kirsten B Moysich
- Division of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - David D Bowtell
- Department of Research, Cancer Genomics and Genetics, Peter MacCallum Cancer Center, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Brenda Y Hernandez
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Hugh Luk
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Sabine Behrens
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mitul Shah
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Audrey Jung
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Prafull Ghatage
- Department of Oncology, Division of Gynecologic Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jennifer Alsop
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Kathryn Alsop
- Department of Research, Cancer Genomics and Genetics, Peter MacCallum Cancer Center, Melbourne, VIC, Australia
| | - Jesús García-Donas
- Medical Oncology Service, HM Hospitales - Centro Integral Oncológico HM Clara Campal, Madrid, Spain
| | - Pamela J Thompson
- Samuel Oschin Comprehensive Cancer Institute, Cancer Prevention and Genetics Program, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Anthony J Swerdlow
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK.,Division of Breast Cancer Research, The Institute of Cancer Research, London, UK
| | - Chloe Karpinskyj
- Gynaecological Cancer Research Centre, Women's Cancer, Institute for Women's Health, University College London, London, UK
| | | | - María J García
- Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.,Biomedical Network on Rare Diseases (CIBERER), Madrid, Spain
| | - Susha Deen
- Department of Histopathology, Queen's Medical Centre, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Lynne R Wilkens
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - José Palacios
- Pathology Department, IRYCIS, CIBERONC, Universidad de Alcalá, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Andrew Berchuck
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, USA
| | | | - James D Brenton
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Linda S Cook
- University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, USA.,Department of Cancer Epidemiology and Prevention Research, Alberta Health Services, Calgary, AB, Canada
| | - Ellen L Goode
- Department of Health Science Research, Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
| | - David G Huntsman
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,British Columbia's Ovarian Cancer Research (OVCARE) Program, Vancouver General Hospital, BC Cancer Agency and University of British Columbia, Vancouver, BC, Canada.,Department of Molecular Oncology, BC Cancer Agency Research Centre, Vancouver, BC, Canada
| | - Susan J Ramus
- School of Women's and Children's Health, Faculty of Medicine, University of NSW Sydney, Sydney, NSW, Australia.,The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Martin Köbel
- Department of Pathology and Laboratory Medicine, University of Calgary, Foothills Medical Center, Calgary, AB, Canada
| |
Collapse
|
7
|
Rambau PF, Vierkant RA, Intermaggio MP, Kelemen LE, Goodman MT, Herpel E, Pharoah PD, Kommoss S, Jimenez-Linan M, Karlan BY, Gentry-Maharaj A, Menon U, Polo SH, Candido Dos Reis FJ, Doherty JA, Gayther SA, Sharma R, Larson MC, Harnett PR, Hatfield E, de Andrade JM, Nelson GS, Steed H, Schildkraut JM, Carney ME, Høgdall E, Whittemore AS, Widschwendter M, Kennedy CJ, Wang F, Wang Q, Wang C, Armasu SM, Daley F, Coulson P, Jones ME, Anglesio MS, Chow C, de Fazio A, García-Closas M, Brucker SY, Cybulski C, Harris HR, Hartkopf AD, Huzarski T, Jensen A, Lubiński J, Oszurek O, Benitez J, Mina F, Staebler A, Taran FA, Pasternak J, Talhouk A, Rossing MA, Hendley J, Edwards RP, Fereday S, Modugno F, Ness RB, Sieh W, El-Bahrawy MA, Winham SJ, Lester J, Kjaer SK, Gronwald J, Sinn P, Fasching PA, Chang-Claude J, Moysich KB, Bowtell DD, Hernandez BY, Luk H, Behrens S, Shah M, Jung A, Ghatage P, Alsop J, Alsop K, García-Donas J, Thompson PJ, Swerdlow AJ, Karpinskyj C, Cazorla-Jiménez A, García MJ, Deen S, Wilkens LR, Palacios J, Berchuck A, Koziak JM, Brenton JD, Cook LS, Goode EL, Huntsman DG, Ramus SJ, Köbel M. Association of p16 expression with prognosis varies across ovarian carcinoma histotypes: an Ovarian Tumor Tissue Analysis consortium study. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2018. [PMID: 30062862 DOI: 10.1002/cjp2.109]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
We aimed to validate the prognostic association of p16 expression in ovarian high-grade serous carcinomas (HGSC) and to explore it in other ovarian carcinoma histotypes. p16 protein expression was assessed by clinical-grade immunohistochemistry in 6525 ovarian carcinomas including 4334 HGSC using tissue microarrays from 24 studies participating in the Ovarian Tumor Tissue Analysis consortium. p16 expression patterns were interpreted as abnormal (either overexpression referred to as block expression or absence) or normal (heterogeneous). CDKN2A (which encodes p16) mRNA expression was also analyzed in a subset (n = 2280) mostly representing HGSC (n = 2010). Association of p16 expression with overall survival (OS) was determined within histotypes as was CDKN2A expression for HGSC only. p16 block expression was most frequent in HGSC (56%) but neither protein nor mRNA expression was associated with OS. However, relative to heterogeneous expression, block expression was associated with shorter OS in endometriosis-associated carcinomas, clear cell [hazard ratio (HR): 2.02, 95% confidence (CI) 1.47-2.77, p < 0.001] and endometrioid (HR: 1.88, 95% CI 1.30-2.75, p = 0.004), while absence was associated with shorter OS in low-grade serous carcinomas (HR: 2.95, 95% CI 1.61-5.38, p = 0.001). Absence was most frequent in mucinous carcinoma (50%), and was not associated with OS in this histotype. The prognostic value of p16 expression is histotype-specific and pattern dependent. We provide definitive evidence against an association of p16 expression with survival in ovarian HGSC as previously suggested. Block expression of p16 in clear cell and endometrioid carcinoma should be further validated as a prognostic marker, and absence in low-grade serous carcinoma justifies CDK4 inhibition.
Collapse
Affiliation(s)
- Peter F Rambau
- Department of Pathology and Laboratory Medicine, University of Calgary, Foothills Medical Center, Calgary, AB, Canada.,Pathology Department, Catholic University of Health and Allied Sciences-Bugando, Mwanza, Tanzania
| | - Robert A Vierkant
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Maria P Intermaggio
- School of Women's and Children's Health, Faculty of Medicine, University of NSW Sydney, Sydney, NSW, Australia
| | - Linda E Kelemen
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Marc T Goodman
- Samuel Oschin Comprehensive Cancer Institute, Cancer Prevention and Genetics Program, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Esther Herpel
- National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany
| | - Paul D Pharoah
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK.,Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Stefan Kommoss
- Department of Women's Health, Tübingen University Hospital, Tübingen, Germany
| | | | - Beth Y Karlan
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Aleksandra Gentry-Maharaj
- Gynaecological Cancer Research Centre, Women's Cancer, Institute for Women's Health, University College London, London, UK
| | - Usha Menon
- Gynaecological Cancer Research Centre, Women's Cancer, Institute for Women's Health, University College London, London, UK
| | - Susanna Hernando Polo
- Medical Oncology Service, Hospital Universitario Funcación Alcorcón, Alcorcón, Spain
| | - Francisco J Candido Dos Reis
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Jennifer Anne Doherty
- Department of Population Health Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Simon A Gayther
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Center for Cancer Prevention and Translational Genomics, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Raghwa Sharma
- Pathology West ICPMR Westmead, Westmead Hospital, The University of Sydney, Sydney, NSW, Australia.,University of Western Sydney at Westmead Hospital, Westmead, NSW, Australia
| | - Melissa C Larson
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Paul R Harnett
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia.,The Crown Princess Mary Cancer Centre Westmead, Sydney-West Cancer Network, Westmead Hospital, Sydney, NSW, Australia
| | - Emma Hatfield
- Department of Pathology and Laboratory Medicine, University of Calgary, Foothills Medical Center, Calgary, AB, Canada
| | - Jurandyr M de Andrade
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Gregg S Nelson
- Department of Oncology, Division of Gynecologic Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Helen Steed
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Royal Alexandra Hospital, Edmonton, AB, Canada
| | - Joellen M Schildkraut
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - Micheal E Carney
- John A. Burns School of Medicine, Department of Obstetrics and Gynecology, University of Hawaii, Honolulu, HI, USA
| | - Estrid Høgdall
- Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark.,Molecular Unit, Department of Pathology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Alice S Whittemore
- Department of Health Research and Policy - Epidemiology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Martin Widschwendter
- Gynaecological Cancer Research Centre, Women's Cancer, Institute for Women's Health, University College London, London, UK
| | - Catherine J Kennedy
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia.,Department of Gynaecological Oncology, Westmead Hospital, Sydney, NSW, Australia
| | - Frances Wang
- Cancer Control and Population Sciences, Duke Cancer Institute, Durham, NC, USA.,Department of Community and Family Medicine, Duke University Medical Center, Durham, NC, USA
| | - Qin Wang
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Chen Wang
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Sebastian M Armasu
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Frances Daley
- Division of Breast Cancer Research, Institute of Cancer Research, London, UK.,Division of Bioscience, Brunel University, London, UK
| | - Penny Coulson
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - Micheal E Jones
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - Micheal S Anglesio
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Christine Chow
- Genetic Pathology Evaluation Centre, Vancouver General Hospital and University of British Columbia, Vancouver, BC, Canada
| | - Anna de Fazio
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia.,Department of Gynaecological Oncology, Westmead Hospital, Sydney, NSW, Australia
| | - Montserrat García-Closas
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK.,Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Sara Y Brucker
- Department of Gynecology and Obstetrics, University of Tübingen, Tübingen, Germany
| | - Cezary Cybulski
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Holly R Harris
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Department of Environmental Medicine, Division of Nutritional Epidemiology, Karolinska Institutet, Stockholm, Sweden
| | - Andreas D Hartkopf
- Department of Women's Health, Tübingen University Hospital, Tübingen, Germany
| | - Tomasz Huzarski
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Allan Jensen
- Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Jan Lubiński
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Oleg Oszurek
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Javier Benitez
- Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.,Biomedical Network on Rare Diseases (CIBERER), Madrid, Spain
| | - Fady Mina
- Department of Pathology and Laboratory Medicine, University of Calgary, Foothills Medical Center, Calgary, AB, Canada
| | - Annette Staebler
- Institute of Pathology, Tübingen University Hospital, Tübingen, Germany
| | - Florin Andrei Taran
- Department of Women's Health, Tübingen University Hospital, Tübingen, Germany
| | - Jana Pasternak
- Department of Women's Health, Tübingen University Hospital, Tübingen, Germany
| | - Aline Talhouk
- British Columbia's Ovarian Cancer Research (OVCARE) Program, Vancouver General Hospital, BC Cancer Agency and University of British Columbia, Vancouver, BC, Canada
| | - Mary Anne Rossing
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Joy Hendley
- Department of Research, Cancer Genomics and Genetics, Peter MacCallum Cancer Center, Melbourne, VIC, Australia
| | -
- Peter MacCallum Cancer Center, Melbourne, VIC, Australia.,Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Robert P Edwards
- Ovarian Cancer Center of Excellence, Womens Cancer Research Program, Magee-Womens Research Institute and University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA.,Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sian Fereday
- Department of Research, Cancer Genomics and Genetics, Peter MacCallum Cancer Center, Melbourne, VIC, Australia
| | - Francesmary Modugno
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Womens Cancer Research Center, Magee-Womens Research Institute and Hillman Cancer Center, Pittsburgh, PA, USA
| | - Roberta B Ness
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Weiva Sieh
- Department of Genetics and Genomic Sciences, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mona A El-Bahrawy
- Department of Histopathology, Imperial College London, Hammersmith Hospital, London, UK
| | - Stacey J Winham
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Jenny Lester
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Susanne K Kjaer
- Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark.,Department of Gynaecology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jacek Gronwald
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Peter Sinn
- Department of Pathology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Peter A Fasching
- David Geffen School of Medicine, Department of Medicine Division of Hematology and Oncology, University of California at Los Angeles, Los Angeles, CA, USA.,Department of Gynecology and Obstetrics, Comprehensive Cancer Center ER-EMN, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Cancer Epidemiology Group, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kirsten B Moysich
- Division of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - David D Bowtell
- Department of Research, Cancer Genomics and Genetics, Peter MacCallum Cancer Center, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Brenda Y Hernandez
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Hugh Luk
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Sabine Behrens
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mitul Shah
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Audrey Jung
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Prafull Ghatage
- Department of Oncology, Division of Gynecologic Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jennifer Alsop
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Kathryn Alsop
- Department of Research, Cancer Genomics and Genetics, Peter MacCallum Cancer Center, Melbourne, VIC, Australia
| | - Jesús García-Donas
- Medical Oncology Service, HM Hospitales - Centro Integral Oncológico HM Clara Campal, Madrid, Spain
| | - Pamela J Thompson
- Samuel Oschin Comprehensive Cancer Institute, Cancer Prevention and Genetics Program, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Anthony J Swerdlow
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK.,Division of Breast Cancer Research, The Institute of Cancer Research, London, UK
| | - Chloe Karpinskyj
- Gynaecological Cancer Research Centre, Women's Cancer, Institute for Women's Health, University College London, London, UK
| | | | - María J García
- Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.,Biomedical Network on Rare Diseases (CIBERER), Madrid, Spain
| | - Susha Deen
- Department of Histopathology, Queen's Medical Centre, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Lynne R Wilkens
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - José Palacios
- Pathology Department, IRYCIS, CIBERONC, Universidad de Alcalá, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Andrew Berchuck
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, USA
| | | | - James D Brenton
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Linda S Cook
- University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, USA.,Department of Cancer Epidemiology and Prevention Research, Alberta Health Services, Calgary, AB, Canada
| | - Ellen L Goode
- Department of Health Science Research, Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
| | - David G Huntsman
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,British Columbia's Ovarian Cancer Research (OVCARE) Program, Vancouver General Hospital, BC Cancer Agency and University of British Columbia, Vancouver, BC, Canada.,Department of Molecular Oncology, BC Cancer Agency Research Centre, Vancouver, BC, Canada
| | - Susan J Ramus
- School of Women's and Children's Health, Faculty of Medicine, University of NSW Sydney, Sydney, NSW, Australia.,The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Martin Köbel
- Department of Pathology and Laboratory Medicine, University of Calgary, Foothills Medical Center, Calgary, AB, Canada
| |
Collapse
|
8
|
Aftab A, Shahzad S, Hussain HMJ, Khan R, Irum S, Tabassum S. CDKN2A/P16INK4A variants association with breast cancer and their in-silico analysis. Breast Cancer 2018; 26:11-28. [DOI: 10.1007/s12282-018-0894-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 07/13/2018] [Indexed: 12/12/2022]
|
9
|
Kwon NH, Lee JY, Ryu YL, Kim C, Kong J, Oh S, Kang BS, Ahn HW, Ahn SG, Jeong J, Kim HK, Kim JH, Han DY, Park MC, Kim D, Takase R, Masuda I, Hou YM, Jang SI, Chang YS, Lee DK, Kim Y, Wang MW, Basappa, Kim S. Stabilization of Cyclin-Dependent Kinase 4 by Methionyl-tRNA Synthetase in p16 INK4a-Negative Cancer. ACS Pharmacol Transl Sci 2018; 1:21-31. [PMID: 32219202 DOI: 10.1021/acsptsci.8b00001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Indexed: 12/23/2022]
Abstract
Although abnormal increases in the level or activity of cyclin-dependent kinase 4 (CDK4) occur frequently in cancer, the underlying mechanism is not fully understood. Here, we show that methionyl-tRNA synthetase (MRS) specifically stabilizes CDK4 by enhancing the formation of the complex between CDK4 and a chaperone protein. Knockdown of MRS reduced the CDK4 level, resulting in G0/G1 cell cycle arrest. The effects of MRS on CDK4 stability were more prominent in the tumor suppressor p16INK4a-negative cancer cells because of the competitive relationship of the two proteins for binding to CDK4. Suppression of MRS reduced cell transformation and the tumorigenic ability of a p16INK4a-negative breast cancer cell line in vivo. Further, the MRS levels showed a positive correlation with those of CDK4 and the downstream signals at high frequency in p16INK4a-negative human breast cancer tissues. This work revealed an unexpected functional connection between the two enzymes involving protein synthesis and the cell cycle.
Collapse
Affiliation(s)
- Nam Hoon Kwon
- Medicinal Bioconvergence Research Center, Seoul National University, Suwon, 16229, Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Korea
| | - Jin Young Lee
- Medicinal Bioconvergence Research Center, Seoul National University, Suwon, 16229, Korea
| | - Ye-Lim Ryu
- Medicinal Bioconvergence Research Center, Seoul National University, Suwon, 16229, Korea
| | - Chanhee Kim
- Medicinal Bioconvergence Research Center, Seoul National University, Suwon, 16229, Korea
| | - Jiwon Kong
- Medicinal Bioconvergence Research Center, Seoul National University, Suwon, 16229, Korea
| | - Seongeun Oh
- Medicinal Bioconvergence Research Center, Seoul National University, Suwon, 16229, Korea
| | - Beom Sik Kang
- School of Life Science and Biotechnology, Kyungpook National University, Daegu, 41566, Korea
| | - Hye Won Ahn
- Medicinal Bioconvergence Research Center, Seoul National University, Suwon, 16229, Korea
| | - Sung Gwe Ahn
- Breast Cancer Center, Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Joon Jeong
- Breast Cancer Center, Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Hoi Kyoung Kim
- Medicinal Bioconvergence Research Center, Seoul National University, Suwon, 16229, Korea
| | - Jong Hyun Kim
- Medicinal Bioconvergence Research Center, Seoul National University, Suwon, 16229, Korea
| | - Dae Young Han
- Medicinal Bioconvergence Research Center, Seoul National University, Suwon, 16229, Korea
| | - Min Chul Park
- Medicinal Bioconvergence Research Center, Seoul National University, Suwon, 16229, Korea
| | - Doyeun Kim
- Medicinal Bioconvergence Research Center, Seoul National University, Suwon, 16229, Korea
| | - Ryuichi Takase
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, United States
| | - Isao Masuda
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, United States
| | - Ya-Ming Hou
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, United States
| | - Sung Ill Jang
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Yoon Soo Chang
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Dong Ki Lee
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Youngeun Kim
- Medicinal Bioconvergence Research Center, Seoul National University, Suwon, 16229, Korea
| | - Ming-Wei Wang
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Basappa
- Laboratory of Chemical Biology, Department of Chemistry, Bangalore University, Palace Road, Bangalore, 560 001, India
| | - Sunghoon Kim
- Medicinal Bioconvergence Research Center, Seoul National University, Suwon, 16229, Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Korea
| |
Collapse
|
10
|
Hormaechea-Agulla D, Gahete MD, Jiménez-Vacas JM, Gómez-Gómez E, Ibáñez-Costa A, L-López F, Rivero-Cortés E, Sarmento-Cabral A, Valero-Rosa J, Carrasco-Valiente J, Sánchez-Sánchez R, Ortega-Salas R, Moreno MM, Tsomaia N, Swanson SM, Culler MD, Requena MJ, Castaño JP, Luque RM. The oncogenic role of the In1-ghrelin splicing variant in prostate cancer aggressiveness. Mol Cancer 2017; 16:146. [PMID: 28851363 PMCID: PMC5576296 DOI: 10.1186/s12943-017-0713-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 08/15/2017] [Indexed: 01/06/2023] Open
Abstract
Background The Ghrelin-system is a complex, pleiotropic family composed of several peptides, including native-ghrelin and its In1-ghrelin splicing variant, and receptors (GHSR 1a/b), which are dysregulated in various endocrine-related tumors, where they associate to pathophysiological features, but the presence, functional role, and mechanisms of actions of In1-ghrelin splicing variant in prostate-cancer (PCa), is completely unexplored. Herein, we aimed to determine the presence of key ghrelin-system components (native-ghrelin, In1-ghrelin, GHSR1a/1b) and their potential pathophysiological role in prostate cancer (PCa). Methods In1-ghrelin and native-ghrelin expression was evaluated by qPCR in prostate tissues from patients with high PCa-risk (n = 52; fresh-tumoral biopsies), and healthy-prostates (n = 12; from cystoprostatectomies) and correlated with clinical parameters using Spearman-test. In addition, In1-ghrelin and native-ghrelin was measured in plasma from an additional cohort of PCa-patients with different risk levels (n = 30) and control-healthy patients (n = 20). In vivo functional (proliferation/migration) and mechanistic (gene expression/signaling-pathways) assays were performed in PCa-cell lines in response to In1-ghrelin and native-ghrelin treatment, overexpression and/or silencing. Finally, tumor progression was monitored in nude-mice injected with PCa-cells overexpressing In1-ghrelin, native-ghrelin and empty vector (control). Results In1-ghrelin, but not native-ghrelin, was overexpressed in high-risk PCa-samples compared to normal-prostate (NP), and this expression correlated with that of PSA. Conversely, GHSR1a/1b expression was virtually absent. Remarkably, plasmatic In1-ghrelin, but not native-ghrelin, levels were also higher in PCa-patients compared to healthy-controls. Furthermore, In1-ghrelin treatment/overexpression, and to a much lesser extent native-ghrelin, increased aggressiveness features (cell-proliferation, migration and PSA secretion) of NP and PCa cells. Consistently, nude-mice injected with PC-3-cells stably-transfected with In1-ghrelin, but not native-ghrelin, presented larger tumors. These effects were likely mediated by ERK1/2-signaling activation and involved altered expression of key oncogenes/tumor suppressor genes. Finally, In1-ghrelin silencing reduced cell-proliferation and PSA secretion from PCa cells. Conclusions Altogether, our results indicate that In1-ghrelin levels (in tissue) and circulating levels (in plasma) are increased in PCa where it can regulate key pathophysiological processes, thus suggesting that In1-ghrelin may represent a novel biomarker and a new therapeutic target in PCa. Electronic supplementary material The online version of this article (doi:10.1186/s12943-017-0713-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Daniel Hormaechea-Agulla
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain.,Reina Sofia University Hospital (HURS), Córdoba, Spain.,CIBERobn, Córdoba, Spain.,ceiA3, Córdoba, Spain
| | - Manuel D Gahete
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain.,Reina Sofia University Hospital (HURS), Córdoba, Spain.,CIBERobn, Córdoba, Spain.,ceiA3, Córdoba, Spain
| | - Juan M Jiménez-Vacas
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain.,Reina Sofia University Hospital (HURS), Córdoba, Spain.,CIBERobn, Córdoba, Spain.,ceiA3, Córdoba, Spain
| | - Enrique Gómez-Gómez
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Reina Sofia University Hospital (HURS), Córdoba, Spain.,Urology Service, HURS/IMIBIC, Córdoba, Spain
| | - Alejandro Ibáñez-Costa
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain.,Reina Sofia University Hospital (HURS), Córdoba, Spain.,CIBERobn, Córdoba, Spain.,ceiA3, Córdoba, Spain
| | - Fernando L-López
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain.,Reina Sofia University Hospital (HURS), Córdoba, Spain.,CIBERobn, Córdoba, Spain.,ceiA3, Córdoba, Spain
| | - Esther Rivero-Cortés
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain.,Reina Sofia University Hospital (HURS), Córdoba, Spain.,CIBERobn, Córdoba, Spain.,ceiA3, Córdoba, Spain
| | - André Sarmento-Cabral
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain.,Reina Sofia University Hospital (HURS), Córdoba, Spain.,CIBERobn, Córdoba, Spain.,ceiA3, Córdoba, Spain
| | - José Valero-Rosa
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Reina Sofia University Hospital (HURS), Córdoba, Spain.,Urology Service, HURS/IMIBIC, Córdoba, Spain
| | - Julia Carrasco-Valiente
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Reina Sofia University Hospital (HURS), Córdoba, Spain.,Urology Service, HURS/IMIBIC, Córdoba, Spain
| | - Rafael Sánchez-Sánchez
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Reina Sofia University Hospital (HURS), Córdoba, Spain.,Anatomical Pathology Service, HURS/IMIBIC, Córdoba, Spain
| | - Rosa Ortega-Salas
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Reina Sofia University Hospital (HURS), Córdoba, Spain.,Anatomical Pathology Service, HURS/IMIBIC, Córdoba, Spain
| | - María M Moreno
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Reina Sofia University Hospital (HURS), Córdoba, Spain.,Anatomical Pathology Service, HURS/IMIBIC, Córdoba, Spain
| | | | - Steve M Swanson
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | | | - María J Requena
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Reina Sofia University Hospital (HURS), Córdoba, Spain.,Urology Service, HURS/IMIBIC, Córdoba, Spain
| | - Justo P Castaño
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain. .,Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain. .,Reina Sofia University Hospital (HURS), Córdoba, Spain. .,CIBERobn, Córdoba, Spain. .,ceiA3, Córdoba, Spain.
| | - Raúl M Luque
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain. .,Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain. .,Reina Sofia University Hospital (HURS), Córdoba, Spain. .,CIBERobn, Córdoba, Spain. .,ceiA3, Córdoba, Spain.
| |
Collapse
|
11
|
Pollack A, Kwon D, Walker G, Khor LY, Horwitz EM, Buyyounouski MK, Stoyanova R. Prospective Validation of Diagnostic Tumor Biomarkers in Men Treated With Radiotherapy for Prostate Cancer. J Natl Cancer Inst 2017; 109:1-8. [PMID: 28376214 DOI: 10.1093/jnci/djw232] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 09/12/2016] [Indexed: 12/20/2022] Open
Abstract
Background In prior retrospective studies, we assessed a number of prostate tumor tissue biomarkers that were associated independently with the clinical outcome of men treated with radiotherapy (RT) ± androgen deprivation therapy (ADT). In this report, the associations of selected biomarkers with biochemical or clinical disease failure (BCDF) were prospectively evaluated in men with T1-T3 prostate cancer on a randomized hypofractionation trial. Methods Biomarkers were analyzed in 263 of 303 men randomly assigned to standard vs moderate hypofractionation. Median follow-up was 65.9 months. Archival tissue was analyzed for Ki-67 (n = 231), MDM2 (n = 209), p16 (n = 195), Cox-2 (n = 126), p53 (n = 206), bcl2 (n = 223), bax (n = 210), and PKA (n = 160). The base model for multivariable Fine-Gray regression analysis included treatment assignment and risk groups. All statistical tests were two-sided. Results Each biomarker was tested one at a time relative to the base model and selected for inclusion in multivariable analysis. Ki-67 (hazard ratio [HR] = 2.31, 95% confidence interval [CI] = 1.19 to 4.48, P = .01) and bcl2&bax (HR = 2.19, 95% CI = 1.08 to 4.46, P = .03) were statistically significantly related to higher BCDF and were independently statistically significant when considered jointly (Ki-67: HR = 2.26, 95% CI = 1.12 to 4.58, P = .02; bcl2&bax: HR = 2.14, 95% CI = 1.03 to 4.41, P = .04). At 2.5 years postradiotherapy, the C-index of Ki-67 was 73.2%, while for the base model was only 46.2%; Ki-67 was the most statistically significant when tested without bcl2&bax. Conclusions In this prospective multiple biomarker analysis in men with prostate cancer treated with RT±ADT, both Ki-67 and bcl2&bax were independently related to early BCDF; however, Ki-67 alone is indicated to be the most clinically meaningful by C-index analysis and is universally available.
Collapse
Affiliation(s)
- Alan Pollack
- Departments of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Deukwoo Kwon
- Biostatistics and Bioinformatics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Gail Walker
- Biostatistics and Bioinformatics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Li Yan Khor
- Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Eric M Horwitz
- Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | | | - Radka Stoyanova
- Departments of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
12
|
Yang CC, Fazli L, Loguercio S, Zharkikh I, Aza-Blanc P, Gleave ME, Wolf DA. Downregulation of c-SRC kinase CSK promotes castration resistant prostate cancer and pinpoints a novel disease subclass. Oncotarget 2016; 6:22060-71. [PMID: 26091350 PMCID: PMC4673146 DOI: 10.18632/oncotarget.4279] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 06/08/2015] [Indexed: 01/03/2023] Open
Abstract
SRC kinase is activated in castration resistant prostate cancer (CRPC), phosphorylates the androgen receptor (AR), and causes its ligand-independent activation as a transcription factor. However, activating SRC mutations are exceedingly rare in human tumors, and mechanisms of ectopic SRC activation therefore remain largely unknown. Performing a functional genomics screen, we found that downregulation of SRC inhibitory kinase CSK is sufficient to overcome growth arrest induced by depriving human prostate cancer cells of androgen. CSK knockdown led to ectopic SRC activation, increased AR signaling, and resistance to anti-androgens. Consistent with the in vitro observations, stable knockdown of CSK conferred castration resistance in mouse xenograft models, while sensitivity to the tyrosine kinase inhibitor dasatinib was retained. Finally, CSK was found downregulated in a distinct subset of CRPCs marked by AR amplification and ETS2 deletion but lacking PTEN and RB1 mutations. These results identify CSK downregulation as a principal driver of SRC activation and castration resistance and validate SRC as a drug target in a molecularly defined subclass of CRPCs.
Collapse
Affiliation(s)
- Chih-Cheng Yang
- Tumor Initiation & Maintenance Program, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA.,Functional Genomics Core, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | - Ladan Fazli
- Vancouver Prostate Centre, Vancouver, BC, Canada V6H 3Z6
| | - Salvatore Loguercio
- San Diego Center for Systems Biology, La Jolla, CA 92037, USA.,Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Irina Zharkikh
- Tumor Analysis Core, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | - Pedro Aza-Blanc
- Functional Genomics Core, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | | | - Dieter A Wolf
- Tumor Initiation & Maintenance Program, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA.,San Diego Center for Systems Biology, La Jolla, CA 92037, USA.,School of Pharmaceutical Sciences & Center for Stress Signaling Networks, Xiamen University, Xiamen 361102, China
| |
Collapse
|
13
|
p16 upregulation is linked to poor prognosis in ERG negative prostate cancer. Tumour Biol 2016; 37:12655-12663. [PMID: 27444279 DOI: 10.1007/s13277-016-5167-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 07/12/2016] [Indexed: 10/21/2022] Open
Abstract
Altered expression of the p16 tumor suppressor is frequently found in prostate cancer, but its role for tumor development and patient prognosis is disputed. In order to clarify the prognostic role of p16 and to draw conclusions on interactions with key molecular features of prostate cancer, we studied p16 expression in a tissue microarray (TMA) with more than 12,400 prostate cancers and attached clinical, pathological, and molecular data such as ERG status and deletions of 3p13, 5q21, 6q15, and PTEN. p16 immunostaining was absent in non-neoplastic prostate cells but was found in 37 % of 9627 interpretable prostate cancers. Finding p16 expression in 58 % of ERG positive but in only 22 % of ERG negative cancers (p < 0.0001), highlights the known androgen-dependence of both genes. Significant associations between p16 upregulation and tumor phenotype or patient prognosis were strictly limited to the subset of ERG negative cancers. For example, p16 positivity increased from 15 % in Gleason ≤3 + 3 to 38 % in Gleason ≥4 + 4 cancers (p < 0.0001) and was associated with early PSA recurrence (p < 0.0001). p16 upregulation was strongly linked to deletions of PTEN (p < 0.0001), highlighting the interaction of both genes in growth control. In conclusion, p16 upregulation is a strong prognostic factor in ERG negative cancers. The strict limitation of its prognostic impact to a molecularly defined subgroup challenges the concept of molecular prognosis testing without considering molecular subtypes.
Collapse
|
14
|
Lynch SM, McKenna MM, Walsh CP, McKenna DJ. miR-24 regulates CDKN1B/p27 expression in prostate cancer. Prostate 2016; 76:637-48. [PMID: 26847530 DOI: 10.1002/pros.23156] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 01/08/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND MicroRNAs (miRNAs) are small, non-coding RNA molecules with an important role in cancer. In prostate cancer, several miRNAs are expressed abnormally suggesting they may be useful markers for diagnosis, prognosis, and potential therapeutic intervention in this disease. However, the contribution of individual miRNAs to the development and progression of this disease remains poorly understood. This study investigated the role of miR-24, which has not been extensively studied in relation to prostate cancer. METHODS We used PCR to investigate the expression of miR-24 in a panel of prostate cancer cell-lines and in a series of clinical prostate biopsy specimens. The biological significance of miR-24 expression in prostate cancer cells was assessed by a series of in vitro bioassays and the effect on proposed targets p27 (CDKN1B) and p16 (CDK2NA) was investigated. RESULTS We showed that miR-24 expression was significantly lower in prostate cancer cell lines compared to a normal prostate epithelial cell line. Decreased expression of miR-24 was also more frequently observed in both needle core and prostatectomy tumor tissue relative to matched normal tissue. Low miR-24 expression correlated with high PSA serum levels and other markers of increased prostate cancer progression. Importantly, over-expression of miR-24 inhibited cell cycle, proliferation, migration, and clonogenic potential of prostate cancer cells, as well as inducing apoptosis. p27 and p16 were confirmed as targets of miR-24 in prostate cancer cells and a significant inverse correlation between miR-24 and p27 was revealed in clinical prostatectomy specimens. CONCLUSIONS These findings provide evidence that miR-24 has a tumor suppressor role in prostate cancer and also targets p27 and p16 in prostate cancer cells. We propose that it may be a useful progression biomarker or focus of therapeutic intervention for this disease.
Collapse
Affiliation(s)
- Seodhna M Lynch
- Biomedical Sciences Research Institute, University of Ulster, Coleraine, Derry, United Kingdom
| | - Michael M McKenna
- Department of Cellular Pathology, Western Health and Social Care Trust, Altnagelvin Area Hospital, Derry, United Kingdom
| | - Colum P Walsh
- Biomedical Sciences Research Institute, University of Ulster, Coleraine, Derry, United Kingdom
| | - Declan J McKenna
- Biomedical Sciences Research Institute, University of Ulster, Coleraine, Derry, United Kingdom
| |
Collapse
|
15
|
Dal Pra A, Locke JA, Borst G, Supiot S, Bristow RG. Mechanistic Insights into Molecular Targeting and Combined Modality Therapy for Aggressive, Localized Prostate Cancer. Front Oncol 2016; 6:24. [PMID: 26909338 PMCID: PMC4754414 DOI: 10.3389/fonc.2016.00024] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Accepted: 01/22/2016] [Indexed: 12/12/2022] Open
Abstract
Radiation therapy (RT) is one of the mainstay treatments for prostate cancer (PCa). The potentially curative approaches can provide satisfactory results for many patients with non-metastatic PCa; however, a considerable number of individuals may present disease recurrence and die from the disease. Exploiting the rich molecular biology of PCa will provide insights into how the most resistant tumor cells can be eradicated to improve treatment outcomes. Important for this biology-driven individualized treatment is a robust selection procedure. The development of predictive biomarkers for RT efficacy is therefore of utmost importance for a clinically exploitable strategy to achieve tumor-specific radiosensitization. This review highlights the current status and possible opportunities in the modulation of four key processes to enhance radiation response in PCa by targeting the: (1) androgen signaling pathway; (2) hypoxic tumor cells and regions; (3) DNA damage response (DDR) pathway; and (4) abnormal extra-/intracell signaling pathways. In addition, we discuss how and which patients should be selected for biomarker-based clinical trials exploiting and validating these targeted treatment strategies with precision RT to improve cure rates in non-indolent, localized PCa.
Collapse
Affiliation(s)
- Alan Dal Pra
- Radiation Medicine Program, Ontario Cancer Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Jennifer A Locke
- Radiation Medicine Program, Ontario Cancer Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Gerben Borst
- Radiation Medicine Program, Ontario Cancer Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Stephane Supiot
- Integrated Center of Oncology (ICO) René Gauducheau , Nantes , France
| | - Robert G Bristow
- Radiation Medicine Program, Ontario Cancer Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
16
|
Wilkins A, Dearnaley D, Somaiah N. Genomic and Histopathological Tissue Biomarkers That Predict Radiotherapy Response in Localised Prostate Cancer. BIOMED RESEARCH INTERNATIONAL 2015; 2015:238757. [PMID: 26504789 PMCID: PMC4609338 DOI: 10.1155/2015/238757] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 05/24/2015] [Indexed: 12/16/2022]
Abstract
Localised prostate cancer, in particular, intermediate risk disease, has varied survival outcomes that cannot be predicted accurately using current clinical risk factors. External beam radiotherapy (EBRT) is one of the standard curative treatment options for localised disease and its efficacy is related to wide ranging aspects of tumour biology. Histopathological techniques including immunohistochemistry and a variety of genomic assays have been used to identify biomarkers of tumour proliferation, cell cycle checkpoints, hypoxia, DNA repair, apoptosis, and androgen synthesis, which predict response to radiotherapy. Global measures of genomic instability also show exciting capacity to predict survival outcomes following EBRT. There is also an urgent clinical need for biomarkers to predict the radiotherapy fraction sensitivity of different prostate tumours and preclinical studies point to possible candidates. Finally, the increased resolution of next generation sequencing (NGS) is likely to enable yet more precise molecular predictions of radiotherapy response and fraction sensitivity.
Collapse
Affiliation(s)
- Anna Wilkins
- Division of Clinical Studies, The Institute of Cancer Research, London SM2 5NG, UK
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SM2 5NG, UK
| | - David Dearnaley
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SM2 5NG, UK
| | - Navita Somaiah
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SM2 5NG, UK
- Division of Cancer Biology, The Institute of Cancer Research, London SM2 5NG, UK
| |
Collapse
|
17
|
Pollack A, Dignam JJ, Diaz DA, Wu Q, Stoyanova R, Bae K, Dicker AP, Sandler H, Hanks GE, Feng FY. A tissue biomarker-based model that identifies patients with a high risk of distant metastasis and differential survival by length of androgen deprivation therapy in RTOG protocol 92-02. Clin Cancer Res 2014; 20:6379-88. [PMID: 25294917 PMCID: PMC4299458 DOI: 10.1158/1078-0432.ccr-14-0075] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE To examine the relationship between the expression of 7 promising apoptotic/cell proliferation proteins (Ki-67, p53, MDM2, bcl-2, bax, p16, and Cox-2) and risk of distant metastasis. EXPERIMENTAL DESIGN RTOG 92-02 compared external beam radiotherapy (EBRT) to approximately 70 Gy + short-term androgen deprivation therapy (STADT) with EBRT + long-term ADT (LTADT). Immunohistochemical analysis was available for ≥4 biomarkers in 616 of 1,521 assessable cases. Biomarkers were evaluated individually and jointly via multivariable modeling of distant metastasis using competing risks hazards regression, adjusting for age, prostate-specific antigen, Gleason score, T stage, and treatment. RESULTS Modeling identified four biomarkers (Ki-67, MDM2, p16 and Cox-2) that were jointly associated with distant metastasis. The c-index was 0.77 for the full model and 0.70 for the model without the biomarkers; a relative improvement of about 10% (likelihood ratio P < 0.001). Subdivision of the patients into quartiles based on predicted distant metastasis risk identified a high-risk group with 10-year distant metastasis risk of 52.5% after EBRT + STADT and 31% with EBRT + LTADT; associated 10-year prostate cancer-specific mortality (PCSM) risks were 45.9% and 14.5% with STADT and LTADT. CONCLUSION Four biomarkers were found to contribute significantly to a model that predicted distant metastasis and identified a subgroup of patients at a particularly high risk of both distant metastasis and PCSM when EBRT + STADT was used. LTADT resulted in significant reductions in distant metastasis and improvements in PCSM, and there was a suggestion of greater importance in the very high risk subgroup.
Collapse
Affiliation(s)
- Alan Pollack
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, Florida.
| | - James J Dignam
- Department of Public Health Sciences, University of Chicago, Chicago, Illinois; and NRG Oncology Statistics and Data Management Center, Philadelphia, Pennsylvania
| | - Dayssy A Diaz
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, Florida
| | - Qian Wu
- Department of Public Health Sciences, University of Chicago, Chicago, Illinois; and NRG Oncology Statistics and Data Management Center, Philadelphia, Pennsylvania
| | - Radka Stoyanova
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, Florida
| | - Kyounghwa Bae
- Early Clinical Biostatistics, Novartis Pharmaceuticals, East Hanover, New Jersey
| | - Adam P Dicker
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Howard Sandler
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Gerald E Hanks
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Felix Y Feng
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
18
|
Thangavel C, Boopathi E, Ciment S, Liu Y, O’Neill R, Sharma A, McMahon SB, Mellert H, Addya S, Ertel A, Birbe R, Fortina P, Dicker AP, Knudsen KE, Den RB. The retinoblastoma tumor suppressor modulates DNA repair and radioresponsiveness. Clin Cancer Res 2014; 20:5468-5482. [PMID: 25165096 PMCID: PMC4216759 DOI: 10.1158/1078-0432.ccr-14-0326] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE Perturbations in the retinoblastoma pathway are over-represented in advanced prostate cancer; retinoblastoma loss promotes bypass of first-line hormone therapy. Conversely, preliminary studies suggested that retinoblastoma-deficient tumors may become sensitized to a subset of DNA-damaging agents. Here, the molecular and in vivo consequence of retinoblastoma status was analyzed in models of clinical relevance. EXPERIMENTAL DESIGN Experimental work was performed with multiple isogenic prostate cancer cell lines (hormone sensitive: LNCaP and LAPC4 cells and hormone resistant C42, 22Rv1 cells; stable knockdown of retinoblastoma using shRNA). Multiple mechanisms were interrogated including cell cycle, apoptosis, and DNA damage repair. Transcriptome analysis was performed, validated, and mechanisms discerned. Cell survival was measured using clonogenic cell survival assay and in vivo analysis was performed in nude mice with human derived tumor xenografts. RESULTS Loss of retinoblastoma enhanced the radioresponsiveness of both hormone-sensitive and castrate-resistant prostate cancer. Hypersensitivity to ionizing radiation was not mediated by cell cycle or p53. Retinoblastoma loss led to alteration in DNA damage repair and activation of the NF-κB pathway and subsequent cellular apoptosis through PLK3. In vivo xenografts of retinoblastoma-deficient tumors exhibited diminished tumor mass, lower PSA kinetics, and decreased tumor growth after treatment with ionizing radiation (P < 0.05). CONCLUSIONS Loss of retinoblastoma confers increased radiosensitivity in prostate cancer. This hypersensitization was mediated by alterations in apoptotic signaling. Combined, these not only provide insight into the molecular consequence of retinoblastoma loss, but also credential retinoblastoma status as a putative biomarker for predicting response to radiotherapy.
Collapse
Affiliation(s)
| | - Ettickan Boopathi
- Department of Surgery, Division of Urology, Glenolden, Pennsylvania, USA
| | - Steve Ciment
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Yi Liu
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Raymond O’Neill
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Ankur Sharma
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Steve B. McMahon
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Hestia Mellert
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Biomedical Graduate Studies, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Molecular, Cellular and Developmental Biology, University of Colorado at Boulder, Colorado, USA
| | - Sankar Addya
- Cancer Genomics, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Adam Ertel
- Cancer Genomics, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Ruth Birbe
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Paolo Fortina
- Cancer Genomics, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Adam P Dicker
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Karen E Knudsen
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Department of Urology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Robert B Den
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
19
|
Gnanapragasam VJ. Molecular markers to guide primary radical treatment selection in localized prostate cancer. Expert Rev Mol Diagn 2014; 14:871-81. [DOI: 10.1586/14737159.2014.936851] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
20
|
Molecular markers for prostate cancer in formalin-fixed paraffin-embedded tissues. BIOMED RESEARCH INTERNATIONAL 2013; 2013:283635. [PMID: 24371818 PMCID: PMC3859157 DOI: 10.1155/2013/283635] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 10/10/2013] [Indexed: 12/14/2022]
Abstract
Prostate cancer (PCa) is the most frequently diagnosed type of cancer in developed countries. The decisive method of diagnosis is based on the results of biopsies, morphologically evaluated to determine the presence or absence of cancer. Although this approach leads to a confident diagnosis in most cases, it can be improved by using the molecular markers present in the tissue. Both miRNAs and proteins are considered excellent candidates for biomarkers in formalin-fixed paraffin-embedded (FFPE) tissues, due to their stability over long periods of time. In the last few years, a concerted effort has been made to develop the necessary tools for their reliable measurement in these types of samples. Furthermore, the use of these kinds of markers may also help in establishing tumor grade and aggressiveness, as well as predicting the possible outcomes in each particular case for the different treatments available. This would aid clinicians in the decision-making process. In this review, we attempt to summarize and discuss the potential use of microRNA and protein profiles in FFPE tissue samples as markers to better predict PCa diagnosis, progression, and response to therapy.
Collapse
|
21
|
Abstract
p16(INK4a), located on chromosome 9p21.3, is lost among a cluster of neighboring tumor suppressor genes. Although it is classically known for its capacity to inhibit cyclin-dependent kinase (CDK) activity, p16(INK4a) is not just a one-trick pony. Long-term p16(INK4a) expression pushes cells to enter senescence, an irreversible cell-cycle arrest that precludes the growth of would-be cancer cells but also contributes to cellular aging. Importantly, loss of p16(INK4a) is one of the most frequent events in human tumors and allows precancerous lesions to bypass senescence. Therefore, precise regulation of p16(INK4a) is essential to tissue homeostasis, maintaining a coordinated balance between tumor suppression and aging. This review outlines the molecular pathways critical for proper p16(INK4a) regulation and emphasizes the indispensable functions of p16(INK4a) in cancer, aging, and human physiology that make this gene special.
Collapse
Affiliation(s)
- Kyle M LaPak
- Biomedical Research Tower, Rm 586, The Ohio State University, 460 W. 12th Avenue, Columbus, OH 43210.
| | | |
Collapse
|
22
|
Personalized radiation therapy and biomarker-driven treatment strategies: a systematic review. Cancer Metastasis Rev 2013; 32:479-92. [DOI: 10.1007/s10555-013-9419-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
23
|
Stumm L, Burkhardt L, Steurer S, Simon R, Adam M, Becker A, Sauter G, Minner S, Schlomm T, Sirma H, Michl U. Strong expression of the neuronal transcription factor FOXP2 is linked to an increased risk of early PSA recurrence in ERG fusion-negative cancers. J Clin Pathol 2013; 66:563-8. [PMID: 23559350 DOI: 10.1136/jclinpath-2012-201335] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
BACKGROUND AND AIMS Transcription factors of the forkhead box P (FOXP1-4) family have been implicated in various human cancer types before. The relevance and role of neuronal transcription factor FOXP2 in prostate cancer is unknown. METHODS A tissue microarray containing samples from more than 11 000 prostate cancers from radical prostatectomy specimens with clinical follow-up data was analysed for FOXP2 expression by immunohistochemistry. FOXP2 data were also compared with pre-existing ERG fusion (by fluorescence in situ hybridisation and immunohistochemistry) and cell proliferation (Ki67 labelling index) data. RESULTS There was a moderate to strong FOXP2 protein expression in basal and secretory cells of normal prostatic glands. As compared with normal cells, FOXP2 expression was lost or reduced in 25% of cancers. Strong FOXP2 expression was linked to advanced tumour stage, high Gleason score, presence of lymph node metastases and early tumour recurrence (p<0.0001; each) in ERG fusion-negative, but not in ERG fusion-positive cancers. High FOXP2 expression was linked to high Ki67 labelling index (p<0.0001) in all cancers irrespective of ERG fusion status. CONCLUSIONS These data demonstrate that similar high FOXP2 protein levels as in normal prostate epithelium exert a 'paradoxical' oncogenic role in 'non fusion-type' prostate cancer. It may be speculated that interaction of FOXP2 with members of pathways that are specifically activated in 'non fusion-type' cancers may be responsible for this phenomenon.
Collapse
Affiliation(s)
- Laura Stumm
- Institute of Pathology, University Medical Center, Hamburg-Eppendorf, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Kachroo N, Gnanapragasam VJ. The role of treatment modality on the utility of predictive tissue biomarkers in clinical prostate cancer: a systematic review. J Cancer Res Clin Oncol 2013; 139:1-24. [PMID: 23187933 DOI: 10.1007/s00432-012-1351-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 11/02/2012] [Indexed: 01/25/2023]
Abstract
BACKGROUND Tissue biomarkers could pivotally improve clinical outcome prediction following prostate cancer therapy. Clinically, prostate cancer is managed by diverse treatment modalities whose individual influence on a biomarker's predictive ability is not well understood and poorly investigated in the literature. OBJECTIVE We conducted a systematic review to assess the predictive value of biomarkers in different treatment contexts in prostate cancer. STUDY METHODOLOGY A literature search was performed using the MeSH headings "prostate neoplasms" and "biological markers". Rigorous selection criteria identified studies correlating expression with clinical outcomes from primary androgen deprivation therapy (ADT), radical prostatectomy and radiotherapy (± neoadjuvant ADT). STUDY RESULTS Of 10,668 studies identified, 481 papers matched initial inclusion criteria. Following rescreening, 384 studies identified 236 individual tissue biomarkers, of which 29 were predictive on multivariate analysis in at least 2 independent cohorts. The majority were only tested in surgical cohorts. Only 8 predictive biomarkers were tested across all 3 treatments with Ki67 identified as universal predictive marker. p16 showed potential for treatment stratification between surgery and radiotherapy but needs further validation in independent studies. CONCLUSIONS Despite years of research, very few tissue biomarkers retain predictive value in independent validation across therapy context. Currently, none have conclusive ability to help treatment selection. Future biomarker research should consider the therapy context and use uniform methodology and evaluation criteria.
Collapse
Affiliation(s)
- Naveen Kachroo
- Translational Prostate Cancer Group, Hutchison MRC Research Centre, University of Cambridge, Hills Road, Cambridge CB2 0XZ, UK
| | | |
Collapse
|
25
|
Heckman MG, Parker AS, Wu KJ, Hilton TW, Ko SJ, Pisansky TM, Schild SE, Khor LY, Hammond EH, Pollack A, Buskirk SJ. Evaluation of MDM2, p16, and p53 staining levels as biomarkers of biochemical recurrence following salvage radiation therapy for recurrent prostate cancer. Prostate 2012; 72:1757-66. [PMID: 22513981 DOI: 10.1002/pros.22528] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2012] [Accepted: 03/24/2012] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND PURPOSE The selection of appropriate candidates for salvage radiation therapy (SRT) to address a rising PSA following radical prostatectomy remains challenging. Herein, we provide the first evaluation of the ability of staining levels of the tumor based biomarkers MDM2, p16, and p53 to aid in prediction of biochemical recurrence (BCR) among men undergoing SRT for recurrent prostate cancer. MATERIAL AND METHODS We identified 152 patients who were treated with SRT between July 1987 and July 2003. Staining levels of MDM2, p16, and p53 in primary tumor samples removed during prostatectomy were detected using monoclonal antibodies and quantified by use of a computer-assisted method. Associations of staining levels with BCR were evaluated using Cox proportional hazards regression models; relative risks (RRs) and 95% confidence intervals (CIs) were estimated. RESULTS Compared to patients with low staining (≤median) as measured by percentage of cells with nuclear staining, there was no significant difference in risk of BCR for patients with high MDM2 staining (RR: 0.90, 95% CI: 0.57-1.45, P = 0.67), high p16 staining (RR: 0.88, 95% CI: 0.54-1.44, P = 0.62), or high p53 staining (RR: 1.33, 95% CI: 0.84-2.11, P = 0.23) in multivariable analysis. These results were consistent when considering alternate percentile cutpoints and alternate quantifications of biomarker staining. CONCLUSIONS Our results provide evidence that MDM2, p16, and p53 staining levels are not useful in the prediction of BCR after SRT. As such, these biomarkers are of little clinical use in the selection of appropriate candidates for SRT.
Collapse
Affiliation(s)
- Michael G Heckman
- Biostatistics Unit, Mayo Clinic Florida, Jacksonville, Florida 32224, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Tran PT, Hales RK, Zeng J, Aziz K, Salih T, Gajula RP, Chettiar S, Gandhi N, Wild AT, Kumar R, Herman JM, Song DY, DeWeese TL. Tissue biomarkers for prostate cancer radiation therapy. Curr Mol Med 2012; 12:772-87. [PMID: 22292443 PMCID: PMC3412203 DOI: 10.2174/156652412800792589] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Revised: 11/10/2011] [Accepted: 12/20/2011] [Indexed: 12/12/2022]
Abstract
Prostate cancer is the most common cancer and second leading cause of cancer deaths among men in the United States. Most men have localized disease diagnosed following an elevated serum prostate specific antigen test for cancer screening purposes. Standard treatment options consist of surgery or definitive radiation therapy directed by clinical factors that are organized into risk stratification groups. Current clinical risk stratification systems are still insufficient to differentiate lethal from indolent disease. Similarly, a subset of men in poor risk groups need to be identified for more aggressive treatment and enrollment into clinical trials. Furthermore, these clinical tools are very limited in revealing information about the biologic pathways driving these different disease phenotypes and do not offer insights for novel treatments which are needed in men with poor-risk disease. We believe molecular biomarkers may serve to bridge these inadequacies of traditional clinical factors opening the door for personalized treatment approaches that would allow tailoring of treatment options to maximize therapeutic outcome. We review the current state of prognostic and predictive tissue-based molecular biomarkers which can be used to direct localized prostate cancer treatment decisions, specifically those implicated with definitive and salvage radiation therapy.
Collapse
Affiliation(s)
- P T Tran
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medicine, 1550 Orleans Street, CRB2, RM 406, Baltimore, MD 21231, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Sharma P, Patel D, Chaudhary J. Id1 and Id3 expression is associated with increasing grade of prostate cancer: Id3 preferentially regulates CDKN1B. Cancer Med 2012; 1:187-97. [PMID: 23342268 PMCID: PMC3544440 DOI: 10.1002/cam4.19] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 06/18/2012] [Accepted: 06/19/2012] [Indexed: 12/22/2022] Open
Abstract
As transcriptional regulators of basic helix-oop-helix (bHLH) transcription and non-bHLH factors, the inhibitor of differentiation (Id1, Id2, Id3, and Id4) proteins play a critical role in coordinated regulation of cell growth, differentiation, tumorigenesis, and angiogenesis. Id1 regulates prostate cancer (PCa) cell proliferation, apoptosis, and androgen independence, but its clinical significance in PCa remains controversial. Moreover, there is lack of evidence on the expression of Id2 and Id3 in PCa progression. In this study we investigated the expression of Id2 and Id3 and reevaluated the expression of Id1 in PCa. We show that increased Id1 and Id3 protein expression is strongly associated with increasing grade of PCa. At the molecular level, we report that silencing either Id1 or Id3 attenuates cell cycle. Although structurally and mechanistically similar, our results show that both these proteins are noncompensatory at least in PCa progression. Moreover, through gene silencing approaches we show that Id1 and Id3 primarily attenuates CDKN1A (p21) and CDKN1B (p27), respectively. We also demonstrate that silencing Id3 alone significantly attenuates proliferation of PCa cells as compared with Id1. We propose that increased Id1 and Id3 expression attenuates all three cyclin-dependent kinase inhibitors (CDKN2B, -1A, and -1B) resulting in a more aggressive PCa phenotype.
Collapse
Affiliation(s)
- Pankaj Sharma
- Department of Biological Sciences, Centre for Cancer Research and Therapeutics Development, Clark Atlanta University, Atlanta, Georgia, 30314, USA
| | | | | |
Collapse
|
28
|
Schiewer MJ, Augello MA, Knudsen KE. The AR dependent cell cycle: mechanisms and cancer relevance. Mol Cell Endocrinol 2012; 352:34-45. [PMID: 21782001 PMCID: PMC3641823 DOI: 10.1016/j.mce.2011.06.033] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 06/08/2011] [Accepted: 06/27/2011] [Indexed: 01/04/2023]
Abstract
Prostate cancer cells are exquisitely dependent on androgen receptor (AR) activity for proliferation and survival. As these functions are critical targets of therapeutic intervention for human disease, it is imperative to delineate the mechanisms by which AR engages the cell cycle engine. More than a decade of research has revealed that elegant intercommunication between AR and the cell cycle machinery governs receptor-dependent cellular proliferation, and that perturbations in this process occur frequently in human disease. Here, AR-cell cycle interplay and associated cancer relevance will be reviewed.
Collapse
Affiliation(s)
- Matthew J. Schiewer
- Kimmel Cancer Center, Thomas Jefferson University, 233 S 10th St., Philadelphia, PA 19107, USA
- Department of Cancer Biology, Thomas Jefferson University, 233 S 10th St., Philadelphia, PA 19107, USA
| | - Michael A. Augello
- Kimmel Cancer Center, Thomas Jefferson University, 233 S 10th St., Philadelphia, PA 19107, USA
- Department of Cancer Biology, Thomas Jefferson University, 233 S 10th St., Philadelphia, PA 19107, USA
| | - Karen E. Knudsen
- Kimmel Cancer Center, Thomas Jefferson University, 233 S 10th St., Philadelphia, PA 19107, USA
- Department of Cancer Biology, Thomas Jefferson University, 233 S 10th St., Philadelphia, PA 19107, USA
- Department of Urology, Thomas Jefferson University, 233 S 10th St., Philadelphia, PA 19107, USA
- Department of Radiation Oncology, Thomas Jefferson University, 233 S 10th St., Philadelphia, PA 19107, USA
- Corresponding author at: Kimmel Cancer Center, Thomas Jefferson University, 233 S 10th St., BLSB 1008, Philadelphia, PA 19107, USA. Tel.: +1 215 503 8574 (office)/+1 215 503 8573 (lab). (K.E. Knudsen)
| |
Collapse
|
29
|
Aparicio A, Den RB, Knudsen KE. Time to stratify? The retinoblastoma protein in castrate-resistant prostate cancer. Nat Rev Urol 2011; 8:562-8. [PMID: 21811228 DOI: 10.1038/nrurol.2011.107] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
It is generally held that the retinoblastoma (RB) tumor suppressor functions in multiple tissues to protect against tumor development. However, preclinical studies and analysis of tumor samples of early disease did not support an important role of RB loss in the origin of prostate cancer. By contrast, recent observations in the clinical setting and subsequent modeling of RB function indicate that the tumor suppressor has specialized roles in controlling androgen receptor expression in prostate cancer, and primarily functions to prevent progression to the castration-resistant stage of disease. Furthermore, preclinical models have now shown that loss of RB expression or functional activity decreases the effectiveness of hormone therapy, yet seems to increase sensitivity to a subset of chemotherapeutic agents. Here, the current state of knowledge regarding the implications of RB loss for prostate cancer progression will be reviewed, and potential opportunities for developing RB as a metric to predict therapeutic response will be considered.
Collapse
Affiliation(s)
- Ana Aparicio
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | | | | |
Collapse
|
30
|
Witkiewicz AK, Knudsen KE, Dicker AP, Knudsen ES. The meaning of p16(ink4a) expression in tumors: functional significance, clinical associations and future developments. Cell Cycle 2011; 10:2497-503. [PMID: 21775818 DOI: 10.4161/cc.10.15.16776] [Citation(s) in RCA: 223] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The CDKN2A gene is a tumor suppressor that encodes the CDK4/6 inhibitor p16(ink4a). Loss of this tumor suppressor contributes to the bypass of critical senescent signals and is associated with progression to malignant disease. However, the high-level expression of p16(ink4a) in tumors is associated with aggressive subtypes of disease, and in certain clinical settings elevated p16(ink4a) expression is an important determinant for disease prognosis and therapeutic response. These seemingly contradictory facets of p16(ink4a) expression have lead to confusion related to the meaning of this tumor suppression in tumor pathobiology. As reviewed here, the alternative expression of p16(ink4a) represents an ideal marker for considering RB-pathway function, tumor heterogeneity, and novel means for directing therapy.
Collapse
|
31
|
Warren MV, Chan WYI, Ridley JM. Analysis of protein biomarkers in human clinical tumor samples: critical aspects to success from tissue acquisition to analysis. Biomark Med 2011; 5:227-48. [DOI: 10.2217/bmm.11.9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
There has been increased interest in the analysis of protein biomarkers in clinical tumor tissues in recent years. Tissue-based biomarker assays can add value and aid decision-making at all stages of drug development, as well as being developed for use as predictive biomarkers and for patient stratification and prognostication in the clinic. However, there must be an awareness of the legal and ethical issues related to the sourcing of human tissue samples. This article also discusses the limits of scope and critical aspects on the successful use of the following tissue-based methods: immunohistochemistry, tissue microarrays and automated image analysis. Future advances in standardization of tissue biobanking methods, immunohistochemistry and quantitative image analysis techniques are also discussed.
Collapse
Affiliation(s)
| | - WY Iris Chan
- Pathology Diagnostics Ltd, St John’s Innovation Centre, Cowley Road, Cambridge, CB4 0WS, UK
| | - John M Ridley
- Pathology Diagnostics Ltd, St John’s Innovation Centre, Cowley Road, Cambridge, CB4 0WS, UK
| |
Collapse
|
32
|
Udayakumar T, Shareef MM, Diaz DA, Ahmed MM, Pollack A. The E2F1/Rb and p53/MDM2 pathways in DNA repair and apoptosis: understanding the crosstalk to develop novel strategies for prostate cancer radiotherapy. Semin Radiat Oncol 2011; 20:258-66. [PMID: 20832018 DOI: 10.1016/j.semradonc.2010.05.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Both the p53- and E2F1-signaling pathways are defective in almost all types of tumors, suggesting very important roles for their signaling networks in regulating the process of tumorigenesis and therapy response. Studies on Radiation Therapy Oncology Group tissue samples have identified aberrant expression of p53, MDM2 (an E3 ubiquitin ligase that targets p53 for proteosomal degradation), and p16 (an upstream regulator of retinoblastoma and hence E2F1 in prostate cancer); abnormal expression of these biomarkers has been associated with clinical outcome after radiotherapy ± androgen deprivation therapy. Although the proapoptotic properties of p53 are well documented, a relatively new aspect of p53 function as an active mediator of prosurvival signaling pathways is now emerging. E2F1 is a transcription factor that possesses both proapoptotic and prosurvival properties. Thus, the role of E2F1 in the process of tumorigenesis versus apoptosis is a contested issue that needs to be resolved. Furthermore, the role of E2F1 in DNA repair is being increasingly recognized. Thus, novel approaches to curb the prosurvival and DNA repair capability of E2F1 while promoting apoptotic function are of interest. In this review, we discuss the challenges involved in targeting the p53/E2F1 pathways and the crosstalk networks, and further propose potential therapeutic strategies for prostate cancer management.
Collapse
Affiliation(s)
- Thirupandiyur Udayakumar
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | | | | | | | | |
Collapse
|
33
|
Molecular circuits of solid tumors: prognostic and predictive tools for bedside use. Nat Rev Clin Oncol 2010; 7:367-80. [PMID: 20551944 DOI: 10.1038/nrclinonc.2010.84] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The explosion of knowledge in cancer biology in the past two decades has led to the identification of specific molecular circuits in solid tumors. These pathways reflect specific abnormalities thought to drive malignant progression. This knowledge has also generated a vast panel of cancer biomarkers although many of these biomarkers lack sufficient research and validation to be used in the clinic. This Review discusses relevant molecular prognostic and/or predictive biomarkers in the six leading tumors with the highest contribution to cancer mortality: breast, lung, colorectal, prostate, pancreatic and ovarian cancer. Each biomarker is described according to its associated clinicopathological presentation and specific associated molecular interactions. Despite only few biomarkers being currently implemented in clinical practice, a new generation of predictors is emerging that could modify the classic organ-based cancer classification (for example, defects in DNA repair, aberrant MAPK signaling and aberrant PI3K/Akt/mTOR signaling). The advent of high-throughput strategies will also probably substitute monobiomarker strategies.
Collapse
|
34
|
Park SJ, Sung WJ, Kim MJ. p16INK4a, PTEN, E-cadherin, Bcl-2 and Ki-67 Expression in Prostate Cancer: Its Relationship with the Metastatic Potential and Known Prognostic Factors. KOREAN JOURNAL OF PATHOLOGY 2010. [DOI: 10.4132/koreanjpathol.2010.44.6.597] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Seok Ju Park
- Department of Pathology, Yeungnam University College of Medicine, Daegu, Korea
| | - Woo Jung Sung
- Department of Pathology, Yeungnam University College of Medicine, Daegu, Korea
| | - Mi Jin Kim
- Department of Pathology, Yeungnam University College of Medicine, Daegu, Korea
| |
Collapse
|
35
|
Abstract
Prostate cancer has a variable clinical outcome and, therefore, there is a clear need for novel molecular markers that are specifically associated with biologically aggressive disease to improve staging and prognostication and also to provide mechanistic information to facilitate treatment selection. Different candidate biomarkers have been identified that are linked to patient prognosis and/or response to specific treatments. Such molecules are involved in diverse cellular processes (including cell cycle regulation, cell death and apoptosis, signal transduction, cell adhesion, and angiogenesis) within which aberrant activity of several regulatory pathways has been seen in prostate cancer. Although the number of molecular markers continues to grow, mainly because of the advent of high-throughput methods, more work needs to be done to develop uniform standards for their characterization to enable comparison of markers across studies. Moreover, a rate-limiting step in the development of molecular markers is large-scale clinical assessment and their evaluation in the context of prediction model improvement. In fact, thus far, only a few studies have tested and demonstrated whether the addition of new biological markers improves the accuracy of standard clinical models (nomograms) in predicting biochemical progression in patients with clinically localized prostate cancer who underwent radical prostatectomy. Cancer 2009;115(13 suppl):3058-67. (c) 2009 American Cancer Society.
Collapse
Affiliation(s)
- Alessia Lopergolo
- Department of Experimental Oncology, National Cancer Institute, Milan, Italy
| | | |
Collapse
|
36
|
Roach M, Waldman F, Pollack A. Predictive models in external beam radiotherapy for clinically localized prostate cancer. Cancer 2009; 115:3112-20. [PMID: 19544539 PMCID: PMC2760302 DOI: 10.1002/cncr.24348] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Predictive models are being used increasingly in effort to allow physician and patient expectations to be aligned with outcomes that are based on available data. Most predictive models for men who receive external beam radiotherapy for clinically localized prostate cancer are based on Gleason score, clinical tumor classification, and prostate-specific antigen (PSA) levels. More sophisticated models also have been developed that incorporate treatment-related variables, such as the dose of radiation and the use of androgen-deprivation therapy. Most of the predictive models applied to prostate cancer were derived using PSA recurrence rates as the major endpoint, but clinical endpoints have been incorporated increasingly into predictive models. Biomarkers also are increasingly being added to predictive models in an effort to strengthen them. The Radiation Therapy Oncology Group (RTOG) has completed studies on a wide range of markers using tissue from 2 phase 3 trials (RTOG 8610 and 9202). To date, preliminary assessments of p53; DNA ploidy; p16/retinoblastoma 1 protein; Ki-67; mouse double-minute p53 binding protein homolog; Bcl-2/Bcl-2-associated X protein; cytosine, adenine, and guanine repeats; cyclooxygenase-2; signal transducer and activator of transcription 3; cytochrome P450 3A4; and protein kinase A have been completed. Although they are not ready for widespread, routine use, there are reasons to believe that future models will combine these markers with traditional pretreatment and treatment-related variables and will improve our ability to predict outcome and select the optimal treatment. Cancer 2009;115(13 suppl):3112-20. (c) 2009 American Cancer Society.
Collapse
Affiliation(s)
- Mack Roach
- Department of Radiation Oncology, University of California at San Francisco, San Francisco, California, USA.
| | | | | |
Collapse
|
37
|
Khor LY, Bae K, Paulus R, Al-Saleem T, Hammond ME, Grignon DJ, Che M, Venkatesan V, Byhardt RW, Rotman M, Hanks GE, Sandler HM, Pollack A. MDM2 and Ki-67 predict for distant metastasis and mortality in men treated with radiotherapy and androgen deprivation for prostate cancer: RTOG 92-02. J Clin Oncol 2009; 27:3177-84. [PMID: 19470936 DOI: 10.1200/jco.2008.19.8267] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE MDM2 regulates p53, which controls cell cycle arrest and apoptosis. Both proteins, along with Ki-67, which is an established strong determinant of metastasis, have shown promise in predicting the outcome of men treated with radiation therapy (RT) with or without short-term androgen deprivation (STAD). This report compares the utility of abnormal expression of these biomarkers in estimating progression in a cohort of men treated on RTOG 92-02. PATIENTS AND METHODS Adequate tissue for immunohistochemistry was available for p53, Ki-67, and MDM2 analyses in 478 patient cases. The percentage of tumor nuclei staining positive (PSP) was quantified manually or by image analysis, and the per-sample mean intensity score (MIS) was quantified by image analysis. Cox regression models were used to estimate overall mortality (OM), and Fine and Gray's regressions were applied to the end points of distant metastasis (DM) and cause-specific mortality (CSM). Results In multivariate analyses that adjusted for all markers and treatment covariates, MDM2 overexpression was significantly related to DM (P = .02) and OM (P = .003), and Ki-67 overexpression was significantly related to DM (P < .0001), CSM (P = .0007), and OM (P = .01). P53 overexpression was significantly related to OM (P = .02). When considered in combination, the overexpression of both Ki-67 and MDM2 at high levels was associated with significantly increased failure rates for all end points (P < .001 for DM, CSM, and OM). CONCLUSION Combined MDM2 and Ki-67 expression levels were independently related to distant metastasis and mortality and, if validated, could be considered for risk stratification of patients with prostate cancer in clinical trials.
Collapse
Affiliation(s)
- Li-Yan Khor
- Department of Radiation Oncology, University of Miami, Sylvester Comprehensive Cancer Center, 1475 NW 12th Ave, Suite 1501, Miami, FL 33130, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Sirintrapun SJ, Parwani AV. Molecular Pathology of the Genitourinary Tract: Prostate and Bladder. Surg Pathol Clin 2008; 1:211-36. [PMID: 26837907 DOI: 10.1016/j.path.2008.08.002] [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: 11/16/2022]
Abstract
The knowledge of cellular mechanisms in tumors of the prostate and bladder has grown exponentially. Molecular technologies have led to the discovery of TMPRSS2 in prostate cancer and the molecular pathways distinguishing low- and high-grade urothelial neoplasms. UroVysion with fluorescence in situ hybridization is already commonplace as an adjunct to cytologic diagnosis of urothelial neoplasms. This trend portends the future in which classification and diagnosis of tumors of the prostate and bladder through morphologic analysis will be supplemented by molecular information correlating with prognosis and targeted therapy. This article outlines tumor molecular pathology of the prostate and bladder encompassing current genomic, epigenomic, and proteonomic findings.
Collapse
Affiliation(s)
- S Joseph Sirintrapun
- Pathology Informatics, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Anil V Parwani
- Department of Pathology, University of Pittsburgh Medical Center Shadyside Hospital, Room WG 07, 5230 Centre Avenue, Pittsburgh, PA 15232, USA.
| |
Collapse
|
39
|
Abstract
The probability of extraprostatic disease may be estimated based on clinical T-stage, pretreatment prostatic-specific antigen, Gleason score, and percent positive core biopsies. Patients with disease confined to the prostate may be treated with either prostatectomy or radiotherapy (RT). Patients with extraprostatic disease without evidence of distant metastases are best managed with RT. RT consisting of either external beam and/or brachytherapy results in a relatively high likelihood of cure, particularly for those with low- and intermediate-risk disease. The impact of elective nodal RT on survival is unclear. Dose escalation results in improved biochemical relapse-free survival compared with standard dose RT. Androgen deprivation therapy likely improves the probability of disease control in patients with high-risk cancers.
Collapse
|
40
|
Abstract
The retinoblastoma tumour suppressor (RB) is a crucial regulator of cell-cycle progression that is invoked in response to a myriad of anti-mitogenic signals. It has been hypothesized that perturbations of the RB pathway confer a synonymous proliferative advantage to tumour cells; however, recent findings demonstrate context-specific outcomes associated with such lesions. Particularly, loss of RB function is associated with differential response to wide-ranging therapeutic agents. Thus, the status of this tumour suppressor may be particularly informative in directing treatment regimens.
Collapse
Affiliation(s)
- Erik S. Knudsen
- Department of Cancer Biology, Kimmel Cancer Center, Bluemle Life Science Building-Room 1002, 233, South 10th Street, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA.
| | - Karen E. Knudsen
- Department of Cancer Biology, Kimmel Cancer Center, Bluemle Life Science Building-Room 1002, 233, South 10th Street, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
- Department of Urology, Kimmel Cancer Center, Bluemle Life Science Building-Room 1002, 233, South 10th Street, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA.
| |
Collapse
|
41
|
Celebrating 40 Years of Clinical Inquiry: The RTOG'S Ruby Anniversary. Int J Radiat Oncol Biol Phys 2008; 71:321-3. [DOI: 10.1016/j.ijrobp.2008.03.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2008] [Accepted: 03/28/2008] [Indexed: 11/22/2022]
|