1
|
Bakaloudi DR, Talukder R, Enright T, Leary JB, Makrakis D, Diamantopoulos LN, Hobeika C, Thomas VM, Swami U, Zakopoulou R, Bamias A, Brown JR, Pinato DJ, Latchford C, Jindal T, Koshkin VS, Murgić J, Miletić M, Frobe A, Johnson J, Zakharia Y, Alva A, Nguyen CB, Hui G, Drakaki A, Rodriguez-Vida A, Rey-Cárdenas M, Castellano D, Buznego LA, Duran I, Barrera RM, Marmolejo D, McKay RR, Stewart TF, Barata P, Epstein IB, Bellmunt J, Yu EY, Raychaudhuri R, Nadal R, Vakar-Lopez F, Gupta S, Wright JL, Khaki AR, Grivas P. Response and Survival With Immune Checkpoint Inhibitor in Patients With Advanced Urothelial Carcinoma and Histology Subtypes. Clin Genitourin Cancer 2025; 23:102356. [PMID: 40378559 DOI: 10.1016/j.clgc.2025.102356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 05/19/2025]
Abstract
BACKGROUND Immune Checkpoint Inhibitors (ICIs) are used for advanced urothelial carcinoma (aUC) in different settings. Most patients have pure UC (PUC) but about one-third have UC mixed with histology subtypes (HS). We examined outcomes in patients with HS aUC treated with ICI. MATERIALS AND METHODS We included patients from 26 centers with PUC and any HS treated with ICI as 1st line (1L) upfront, maintenance avelumab (mAV), and ≥2nd line [2+L] therapy. We calculated overall and progression-free survival (OS, PFS) and observed response rate (ORR) from ICI start. RESULTS We included 1511 patients; 752 1L, 609 2+L, 150 mAV. 1L: median OS was 15 (95% CI, 12-17) months for patients with PUC (n = 518), 15 (95% CI, 8-23) months for squamous UC (n = 85) (HR = 1.2, [95% CI, 0.8-1.6]), 11 (95% CI, 6-17) months for micropapillary UC (n = 46) (HR = 1.2, [95% 0.8-1.8]), and 21 (95% CI, 12-30) months in patients with UC mixed with ≥2 HS (n = 30), (HR = 0.9, [95% CI, 0.5-1.4]). 2+L: median OS was 9 (95% CI, 8-10) months for patients with PUC (n = 441), 9 (95% CI, 1-12) months for squamous UC (n = 60) (HR = 1.1, (95% CI, 0.8-1.6]), 6 (95% CI, 1-11) months for micropapillary UC (n = 37) (HR = 1.1, [95% 0.7-1.6]), and 7 (95% CI, 4-10) months in patients with UC mixed with ≥2 HS (n = 17), (HR = 1.6, [95% CI, 0.9-3.1]). CONCLUSION We found no significant OS difference between PUC and HS in patients with aUC treated with ICI monotherapy. Limitations include retrospective design, small sample size in several subsets, lack of randomization, no central imaging or pathology review, selection and confounding biases. Results are hypothesis-generating and need prospective validation.
Collapse
Affiliation(s)
| | - Rafee Talukder
- Department of Medicine, Division of Hematology/Oncology, Baylor College of Medicine, Houston, TX
| | - Thomas Enright
- Department of Medicine, University of Washington, Seattle, WA
| | - Jacob B Leary
- Department of Medicine, University of Washington, Seattle, WA
| | - Dimitrios Makrakis
- Department of Medicine, Jacobi Medical Center-Albert Einstein College of Medicine, Bronx, NY
| | | | - Charbel Hobeika
- Department of Hematology and Oncology, Taussig cancer Institute, Cleveland Clinic Foundation, Cleveland, OH
| | - Vinay Mathew Thomas
- Division of Oncology, Department of Medicine, University of Utah, Salt Lake City, UT
| | - Umang Swami
- Division of Oncology, Department of Medicine, University of Utah, Salt Lake City, UT
| | - Roubini Zakopoulou
- 2nd Propaedeutic Dept of Internal Medicine, ATTIKON University Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Aristotelis Bamias
- 2nd Propaedeutic Dept of Internal Medicine, ATTIKON University Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Jason R Brown
- Division of Solid Tumor Oncology, University Hospitals Cleveland Medical Center, Case Western Reserve University Cleveland, OH
| | - David J Pinato
- Department of Surgery and Cancer, Imperial College London, London, UK; Division of Oncology, Department of Translational Medicine (DIMET), University of Piemonte Orientale, Novara, Italy
| | - Charles Latchford
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Tanya Jindal
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, Helen Diller Family Cancer Center, San Francisco, CA
| | - Vadim S Koshkin
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, Helen Diller Family Cancer Center, San Francisco, CA
| | - Jure Murgić
- Department of Oncology and Nuclear Medicine, University Hospital Center Sestre Milosrdnice, Zagreb, Croatia
| | - Marija Miletić
- Department of Oncology and Nuclear Medicine, University Hospital Center Sestre Milosrdnice, Zagreb, Croatia; Department of Medical Oncology, The Royal Marsden NHS Foundation Trust, London, UK
| | - Ana Frobe
- Department of Oncology and Nuclear Medicine, Faculty of Dentistry, University Hospital Center Sestre Milosrdnice, Zagreb, Croatia
| | - Jeffrey Johnson
- Division of Oncology, Department of Medicine, University of Iowa, Iowa City, IA
| | - Yousef Zakharia
- Department of Hematology and Oncology, Mayo Clinic Arizona, Phoenix, AZ
| | - Ajjai Alva
- Division of Oncology, Department of Medicine, University of Michigan, Ann Arbor, MI
| | - Charles B Nguyen
- Division of Oncology, Department of Medicine, University of Michigan, Ann Arbor, MI
| | - Gavin Hui
- Division of Hematology/Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Alexandra Drakaki
- Division of Hematology/Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Alejo Rodriguez-Vida
- Medical Oncology Department, Hospital del Mar, IMM Research Institute, Barcelona, Spain
| | - Macarena Rey-Cárdenas
- Department of Medical Oncology, Gustave Roussy, Paris Saclay University, Villejuif, France
| | - Daniel Castellano
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Lucia Alonso Buznego
- Department of Oncology, University Hospital Marqués of Valdecilla, IDIVAL Santander, Cantabria, Spain
| | - Ignacio Duran
- Department of Oncology, University Hospital Marqués of Valdecilla, IDIVAL Santander, Cantabria, Spain
| | - Rafael Morales Barrera
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Vall d' Hebron University Hospital, Barcelona, Spain
| | - David Marmolejo
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Vall d' Hebron University Hospital, Barcelona, Spain
| | - Rana R McKay
- Moores Cancer Center, University of California San Diego, La Jolla, CA
| | - Tyler F Stewart
- Moores Cancer Center, University of California San Diego, La Jolla, CA
| | - Pedro Barata
- Division of Solid Tumor Oncology, University Hospitals Cleveland Medical Center, Case Western Reserve University Cleveland, OH
| | - Ilana B Epstein
- Dana-Farber Cancer Institute/Brigham and Women's Hospital Harvard Medical School, Boston, MA
| | - Joaquim Bellmunt
- Dana-Farber Cancer Institute/Brigham and Women's Hospital Harvard Medical School, Boston, MA
| | - Evan Y Yu
- Division of Hematology Oncology, Department of Medicine, University of Washington, Seattle, WA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Ruben Raychaudhuri
- Division of Hematology Oncology, Department of Medicine, University of Washington, Seattle, WA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Rosa Nadal
- Division of Hematology Oncology, Department of Medicine, University of Washington, Seattle, WA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Funda Vakar-Lopez
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| | - Shilpa Gupta
- Department of Hematology and Oncology, Taussig cancer Institute, Cleveland Clinic Foundation, Cleveland, OH
| | | | - Ali Raza Khaki
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA.
| | - Petros Grivas
- Division of Hematology Oncology, Department of Medicine, University of Washington, Seattle, WA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA.
| |
Collapse
|
2
|
Chen JF, Al-Ahmadie H. Molecular Classification of Urothelial Carcinoma. Surg Pathol Clin 2025; 18:41-51. [PMID: 39890308 DOI: 10.1016/j.path.2024.09.003] [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] [Indexed: 02/03/2025]
Abstract
Urothelial carcinoma exhibits a wide spectrum of morphologic and molecular heterogeneity. Advances in molecular testing have improved our understanding of the molecular biology of urothelial carcinoma, including recurrent genomic alterations and transcriptomic features, leading to the development of molecular classification schemes with potential therapeutic implications. This review summarizes the molecular characteristics of urothelial carcinoma, focusing on genomic and transcriptomic features, updates on variant histology, and novel biomarkers that may guide contemporary and future clinical management.
Collapse
Affiliation(s)
- Jie-Fu Chen
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center
| | - Hikmat Al-Ahmadie
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center.
| |
Collapse
|
3
|
Lobo A, Collins K, Kaushal S, Acosta AM, Akgul M, Adhya AK, Al-Ahmadie HA, Al-Obaidy KI, Amin A, Amin MB, Aron M, Balzer BL, Biswal R, Mohanty S, Browning L, Chakrabarti I, Cima L, Cimadamore A, Desai S, Dhillon J, Deshwal A, Diego GG, Diwaker P, Galea LA, Magi-Galluzzi C, Giannico GA, Gupta NS, Haider A, Hirsch MS, Iczkowski KA, Arora S, Jain E, Jain D, Jha S, Kandukuri S, Kao CS, Kryvenko ON, Kumar RM, Kumari N, Kunju LP, Kuthi L, Lobo J, Lopez JI, Luthringer DJ, Maclean F, Manini C, Mannan R, Martos MG, Mehra R, Menon S, Mishra P, Moch H, Montironi R, Baisakh MR, Netto GJ, Nigam LK, Osunkoya AO, Pagliuca F, Paner GP, Panizo A, Parwani AV, Picken MM, Prendeville S, Przybycin CG, Purkait S, Queipo FJ, Rao BV, Rao P, Reuter VE, Sancheti S, Sangoi AR, Sardana R, Satturwar S, Shah RB, Sharma S, Dixit M, Verma M, Sirohi D, Smith SC, Soni S, Sundaram S, Swain M, Tretiakova M, Trpkov K, MuñizUnamunzaga G, Zhou M, Williamson SR, Lopez-Beltran A, Cheng L, Mohanty SK. Advances, recognition, and interpretation of molecular heterogeneity among conventional and subtype histology of urothelial carcinoma (UC): a survey among urologic pathologists and comprehensive review of the literature. Histopathology 2024; 85:748-759. [PMID: 39075659 DOI: 10.1111/his.15287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/31/2024]
Abstract
AIMS Urothelial carcinoma (UC) demonstrates significant molecular and histologic heterogeneity. The WHO 2022 classification has hinted at adding molecular signatures to the morphologic diagnosis. As morphology and associated molecular repertoire may potentially translate to choices of and response to therapy and relapse rate, broader acceptability of recognizing these key features among uropathologists is needed. This prompted an international survey to ascertain the practice patterns in classical/subtype UC among uropathologists across the globe. METHODS AND RESULTS A survey instrument was shared among 98 uropathologists using SurveyMonkey software. Anonymized respondent data were analysed. The response rate was 85%. A majority were in concordance with the profiles of luminal (93%) and basal (82%) types. Opinion on the FGFR3 testing platform was variable. While 95% concurred that TERT promoter mutation is the key driver in UC, 72% had the opinion that APOBEC mutagenesis is the main signature in muscle invasive bladder cancer (MIBC). Uropathologists have divergent opinions on MIBC and ERCC2 mutations. Among the participants, 94% would quantify aggressive micropapillary and sarcomatoid histology, while 88% would reevaluate another transurethral resection of the bladder tumour specimen in nonmuscle invasive tumour with micropapillary, small cell, or sarcomatoid histology. A leading number agreed to specific molecular signatures of micropapillary (93%), plasmacytoid (97%), and small cell (86%) subtypes. Ninety-six percent of participants agreed that a small-cell component portends a more aggressive course and should be treated with neoadjuvant chemotherapy and 63% would perform HER2/neu testing only on oncologist's request in advanced tumours. Ninety percent agreed that microsatellite instability testing, although not a standard protocol, should be considered in young patients with upper tract UC. Eighty-six percent agreed that UC with high tumour mutational burden would be a better candidate for immunotherapy. CONCLUSION In the era of precision medicine, enhanced understanding of molecular heterogeneity of UC will contribute to better therapeutic options, novel biomarker discovery, innovative management protocols, and outcomes. Our survey provides a broad perspective of pathologists' perceptions and experience regarding incorporation of histomolecular approaches to "personalize" therapy. Due to variable clinical adoption, there is a need for additional data using uniform study criteria. This will drive generation of best practice guidelines in this area for widespread and consistent clinical utility.
Collapse
Affiliation(s)
- Anandi Lobo
- Department of Pathology, Kapoor Centre of Urology and Pathology, Raipur, India
| | - Katrina Collins
- Department of Pathology, Indiana University Health, Indiana, USA
| | - Seema Kaushal
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Andres M Acosta
- Department of Pathology, Indiana University Health, Indiana, USA
| | - Mahmut Akgul
- Department of Pathology, Albany Medical Center, Albany, USA
| | - Amit K Adhya
- Department of Pathology, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Hikmat A Al-Ahmadie
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, USA
| | | | - Ali Amin
- Department of Pathology, Alpert Medical School of Brown University, Providence, USA
| | - Mahul B Amin
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, USA
| | - Manju Aron
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, USA
| | - Bonnie L Balzer
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Rupanita Biswal
- Department of Pathology, Bagchi Sri Shankara Cancer Hospital, Bhubaneswar, India
| | - Subashish Mohanty
- Department of Pathology, SUM Ultimate Medicare Hospital, Bhubaneswar, India
| | - Lisa Browning
- Department of Pathology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Indranil Chakrabarti
- Department of Pathology, All India Institute of Medical Sciences, Kalyani, India
| | - Luca Cima
- Department of Pathology, Santa Chiara Hospital of Trento, Trento, Italy
| | - Alessia Cimadamore
- Department of Pathology, Molecular Medicine and Cell Therapy Foundation, c/o Polytechnic University of the Marche Region, Ancona, Italy
| | - Sangeeta Desai
- Department of Pathology, Tata Memorial Hospital, Mumbai, India
| | | | | | - Guillermo G Diego
- Department of Pathology, University Gregorio Marañon Hospital, Madrid, Spain
| | - Preeti Diwaker
- Department of Pathology, University College of Medical Sciences, New Delhi, India
| | - Laurence A Galea
- Department of Pathology, Melbourne Pathology, Melbourne, Australia
| | | | | | - Nilesh S Gupta
- Department of Pathology, Henry Ford Health System, Detroit, USA
| | - Aiman Haider
- Department of Pathology, University College London Hospitals NHS Foundation Trust, London, UK
| | | | | | - Samriti Arora
- Department of Pathology, CORE Diagnostics, Gurgaon, India
| | - Ekta Jain
- Department of Pathology, CORE Diagnostics, Gurgaon, India
| | - Deepika Jain
- Department of Pathology, CORE Diagnostics, Gurgaon, India
| | - Shilpy Jha
- Department of Pathology, Advanced Medical and Research Institute, Bhubaneswar, India
| | - Shivani Kandukuri
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, USA
| | - Chia-Sui Kao
- Department of Pathology, Cleveland Clinic, Cleveland, USA
| | - Oleksandr N Kryvenko
- Department of Pathology, University of Miami Miller School of Medicine, Miami, USA
| | - Ramani M Kumar
- Department of Pathology, Dane Diagnostics, Palakkad, India
| | - Niraj Kumari
- Department of Pathology, All India Institute of Medical Sciences, Raebareli, India
| | - Lakshmi P Kunju
- Department of Pathology, University of Michigan, Ann Arbor, USA
| | - Levente Kuthi
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - João Lobo
- Department of Pathology, Portuguese Oncology Institute - Porto, Porto, Portugal
| | - Jose I Lopez
- Department of Pathology, Cruces University Hospital, Barakaldo, Spain
| | | | - Fiona Maclean
- Department of Pathology, Douglass Hanly Moir Pathology, Sydney, Australia
| | - Claudia Manini
- Department of Pathology, University of Turin, Turin, Italy
| | - Rahul Mannan
- Department of Pathology, University of Michigan, Ann Arbor, USA
| | - María G Martos
- Department of Pathology, University Gregorio Marañon Hospital, Madrid, Spain
| | - Rohit Mehra
- Department of Pathology, University of Michigan, Ann Arbor, USA
| | - Santosh Menon
- Department of Pathology, Tata Memorial Hospital, Mumbai, India
| | - Pritinanda Mishra
- Department of Pathology, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Holger Moch
- Department of Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Rodolfo Montironi
- Department of Pathology, Molecular Medicine and Cell Therapy Foundation, c/o Polytechnic University of the Marche Region, Ancona, Italy
| | - Manas R Baisakh
- Department of Pathology, Prolife Diagnostics, Bhubaneswar, India
| | - George J Netto
- Department of Pathology, University of Pennsylvania, Philadelphia, USA
| | - Lovelesh K Nigam
- Department of Pathology, Institute of Kidney Diseases and Research Center, Ahmedabad, India
| | - Adeboye O Osunkoya
- Department of Pathology, Emory University School of Medicine, Atlanta, USA
| | - Francesca Pagliuca
- Department of Pathology, Università degliStudidella Campania Luigi Vanvitelli, Caserta, Italy
| | - Gladell P Paner
- Department of Pathology, University of Chicago, Chicago, USA
| | - Angel Panizo
- Department of Pathology, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - Anil V Parwani
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, USA
| | - Maria M Picken
- Department of Pathology, Loyola University Medical Center, Hines, USA
| | - Susan Prendeville
- Department of Pathology, University Health Network, University of Toronto, Toronto, Canada
| | | | - Suvendu Purkait
- Department of Pathology, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Francisco J Queipo
- Department of Pathology, Hospital Universitario de A Coruna, A Coruna, Spain
| | - B Vishal Rao
- Department of Pathology, Basavatarakam Indo-American Cancer Hospital and Research Institute, Hyderabad, India
| | - Priya Rao
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Victor E Reuter
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, USA
| | - Sankalp Sancheti
- Department of Pathology, Homi Bhabha Cancer Hospital, Punjab, India
| | - Ankur R Sangoi
- Department of Pathology, Stanford University, Stanford, USA
| | - Rohan Sardana
- Department of Pathology, Sardana Laboratories, Jalandhar, India
| | - Swati Satturwar
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, USA
| | - Rajal B Shah
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, USA
| | - Shivani Sharma
- Department of Pathology, CORE Diagnostics, Gurgaon, India
| | - Mallika Dixit
- Department of Pathology, CORE Diagnostics, Gurgaon, India
| | - Monica Verma
- Department of Pathology, CORE Diagnostics, Gurgaon, India
| | - Deepika Sirohi
- Department of Pathology, University of California, San Francisco, USA
| | - Steven C Smith
- Department of Pathology, Virginia Commonwealth University School of Medicine, Richmond, USA
| | - Shailesh Soni
- Department of Pathology, Muljibhai Patel Urological Hospital, Nadiad, India
| | - Sandhya Sundaram
- Department of Pathology, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | | | | | - Kiril Trpkov
- Department of Pathology, University of Calgary, Calgary, Canada
| | | | - Ming Zhou
- Department of Pathology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | | | - Antonio Lopez-Beltran
- Department of Pathology, Unit of Anatomical Pathology, Faculty of Medicine, Cordoba University, Cordoba, Spain
| | - Liang Cheng
- Department of Pathology, Alpert Medical School of Brown University, Providence, USA
| | - Sambit K Mohanty
- Department of Pathology, CORE Diagnostics, Gurgaon, India
- Department of Pathology, Advanced Medical and Research Institute, Bhubaneswar, India
| |
Collapse
|
4
|
Al-Ahmadie H, Netto GJ. Molecular Pathology of Urothelial Carcinoma. Clin Lab Med 2024; 44:181-198. [PMID: 38821640 DOI: 10.1016/j.cll.2023.08.010] [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] [Indexed: 06/02/2024]
Abstract
Urothelial carcinoma is characterized by the presence of a wide spectrum of histopathologic features and molecular alterations that contribute to its morphologic and genomic heterogeneity. It typically harbors high rates of somatic mutations with considerable genomic and transcriptional complexity and heterogeneity that is reflective of its varied histomorphologic and clinical features. This review provides an update on the recent advances in the molecular characterization and novel molecular taxonomy of urothelial carcinoma and variant histologies.
Collapse
Affiliation(s)
- Hikmat Al-Ahmadie
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NW 10065, USA.
| | - George J Netto
- Department of Pathology, University of Alabama at Birmingham, University of Alabama at Birmingham School of Medicine, WP Building, Suite P230, 619 19th Street South, Birmingham, AL 35249-7331, USA.
| |
Collapse
|
5
|
Zheng L, Chen H, Zhao J, Roy-Chowdhuri S, Kamat AM, Alhalabi O, Gao J, Siefker-Radtke A, Hansel DE, Czerniak B, Guo CC. Plasmacytoid urothelial carcinoma of the urinary bladder-A clinicopathological and molecular analysis of 52 cases. Hum Pathol 2024; 148:1-6. [PMID: 38679207 PMCID: PMC11755388 DOI: 10.1016/j.humpath.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/20/2024] [Accepted: 04/22/2024] [Indexed: 05/01/2024]
Abstract
Plasmacytoid urothelial carcinoma (UC) is a rare histologic subtype of bladder cancer that is associated with an aggressive clinical behavior. We analyzed the clinicopathologic and molecular features of plasmacytoid UC in 52 patients from a single institute. The patients included 44 men and 8 women, with a mean age of 64 years (range, 41-91 years). All bladder cancers were high-grade UC, and plasmacytoid component accounted for a mean of 47% of bladder tumors (range, 5-100%). Distinct gene mutations were found in most plasmacytoid UCs (n = 49); the most common mutations were TP53 (n = 30), followed by TERT (n = 20), and CDH1 (n = 18). Copy number analysis was performed in 34 patients, and 13 of them showed copy number variations. Expression of HER2 was analyzed in 18 patients by immunohistochemistry, and 3 of them showed HER2 overexpression, which was confirmed by fluorescence in situ hybridization analysis. Thirty-two patients died of disease in a median of 15 months (range, 1-45 months). No individual gene mutations were significantly associated with clinical outcome, but mutations in the mammalian target of rapamycin (mTOR) pathway, including PICK3CA and PIK3R1 mutations, were associated with a significantly shorter survival duration (p < 0.05). Plasmacytoid UC is an aggressive histologic subtype that demonstrates frequent somatic gene mutations and CNVs, which may underlie its oncogenesis and progression. Gene mutations of the mTOR pathway are associated with poor outcome in a subset of patients with plasmacytoid UC.
Collapse
Affiliation(s)
- Lan Zheng
- The University of Texas MD Anderson Cancer Center, Department of Pathology, Houston, TX, USA
| | - Hui Chen
- The University of Texas MD Anderson Cancer Center, Department of Pathology, Houston, TX, USA
| | - Jianping Zhao
- The University of Texas MD Anderson Cancer Center, Department of Pathology, Houston, TX, USA
| | - Sinchita Roy-Chowdhuri
- The University of Texas MD Anderson Cancer Center, Department of Pathology, Houston, TX, USA
| | - Ashish M Kamat
- The University of Texas MD Anderson Cancer Center, Department of Urology, Houston, TX, USA
| | - Omar Alhalabi
- The University of Texas MD Anderson Cancer Center, Department of Genitourinary Medical Oncology, Houston, TX, USA
| | - Jianjun Gao
- The University of Texas MD Anderson Cancer Center, Department of Genitourinary Medical Oncology, Houston, TX, USA
| | - Arlene Siefker-Radtke
- The University of Texas MD Anderson Cancer Center, Department of Genitourinary Medical Oncology, Houston, TX, USA
| | - Donna E Hansel
- The University of Texas MD Anderson Cancer Center, Department of Pathology, Houston, TX, USA
| | - Bogdan Czerniak
- The University of Texas MD Anderson Cancer Center, Department of Pathology, Houston, TX, USA
| | - Charles C Guo
- The University of Texas MD Anderson Cancer Center, Department of Pathology, Houston, TX, USA.
| |
Collapse
|
6
|
Leary JB, Enright T, Bakaloudi DR, Basnet A, Bratslavsky G, Jacob J, Spiess PE, Li R, Necchi A, Kamat AM, Pavlick DC, Danziger N, Huang RSP, Lin DI, Cheng L, Ross J, Talukder R, Grivas P. Frequency and Nature of Genomic Alterations in ERBB2-Altered Urothelial Bladder Cancer. Target Oncol 2024; 19:447-458. [PMID: 38570422 DOI: 10.1007/s11523-024-01056-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Human epidermal growth factor-2 (HER2) overexpression is an oncogenic driver in many solid tumors, including urothelial bladder cancer (UBC). In addition, activating mutations in the ERBB2 gene have been shown to play an oncogenic role similar to ERBB2 amplification. OBJECTIVE To describe and compare the frequency and nature of genomic alterations (GA) of ERBB2-altered (mutations, amplification) and ERBB2 wild-type UBC. PATIENTS AND METHODS Using a hybrid capture-based comprehensive profiling assay, 9518 UBC cases were grouped by ERBB2 alteration and evaluated for all classes of genomic alterations (GA), tumor mutational burden (TMB), microsatellite instability (MSI), genome-wide loss of heterozygosity (gLOH), and genomic mutational signature. PD-L1 expression was measured by immunohistochemistry (Dako 22C3). Categorical statistical comparisons were performed using Fisher's exact tests. RESULTS A total of 602 (6.3%) UBC cases featured ERBB2 extracellular domain short variant (SV) GA (ECDmut+), 253 (2.7%) cases featured ERBB2 kinase domain SV GA (KDmut+), 866 (9.1%) cases had ERBB2 amplification (amp+), and 7797 (81.9%) cases were ERBB2 wild-type (wt). European genetic ancestry of ECDmut+ was higher than ERBB2wt. Numerous significant associations were observed when comparing GA by group. Notably among these, CDKN2A/MTAP loss were more frequent in ERBB2wt versus ECDmut+ and amp+. ERBB3 GA were more frequent in ECDmut+ and KDmut+ than ERBB2wt. TERT GA were more frequent in ECDmut+, KDmut+, and amp+ versus ERBB2wt. TOP2A amplification was significantly more common in ECDmut+ and amp+ versus ERBB2wt, and TP53 SV GA were significantly higher in ERBB2 amp+ versus ERBB2wt. Mean TMB levels were significantly higher in ECDmut+, KDmut+, and amp+ than in ERBB2wt. Apolipoprotein B mRNA-editing enzyme, catalytic polypeptides (APOBEC) signature was more frequent in ECDmut+, KDmut+, and amp+ versus ERBB2wt. No significant differences were observed in PD-L1 status between groups, while gLOH-high status was more common in amp+ versus ERBB2wt. MSI-high status was more frequent in KDmut+ versus ERBB2wt, and in ERBB2wt than in amp+. CONCLUSIONS We noted important differences in co-occurring GA in ERBB2-altered (ECDmut+, KDmut+, amp+) versus ERBB2wt UBC, as well as higher mean TMB and higher APOBEC mutational signature in the ERBB2-altered groups. Our results can help refine future clinical trial designs and elucidate possible response and resistance mechanisms for ERBB2-altered UBC.
Collapse
Affiliation(s)
- Jacob B Leary
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Thomas Enright
- Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - Alina Basnet
- SUNY Upstate Medical University, Syracuse, NY, USA
| | | | - Joseph Jacob
- SUNY Upstate Medical University, Syracuse, NY, USA
| | - Philippe E Spiess
- Department of Genitourinary Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Roger Li
- Department of Genitourinary Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Andrea Necchi
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | | | | | | | | | | | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School at Brown University, Providence, RI, USA
- Legoretta Cancer Center at Brown University, Providence, RI, USA
- Lifespan Academic Medical Center, Providence, RI, USA
| | | | | | - Petros Grivas
- Department of Medicine, University of Washington, Seattle, WA, USA.
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| |
Collapse
|
7
|
Shih CH, Lin YH, Luo HL, Sung WW. Antibody-drug conjugates targeting HER2 for the treatment of urothelial carcinoma: potential therapies for HER2-positive urothelial carcinoma. Front Pharmacol 2024; 15:1326296. [PMID: 38572425 PMCID: PMC10987710 DOI: 10.3389/fphar.2024.1326296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 02/07/2024] [Indexed: 04/05/2024] Open
Abstract
Urothelial carcinoma (UC) is a common cancer characterized by high morbidity and mortality rates. Despite advancements in treatment, challenges such as recurrence and low response rates persist. Antibody-drug conjugates (ADCs) have emerged as a promising therapeutic approach for various cancers, although their application in UC is currently limited. This review focuses on recent research regarding ADCs designed to treat UC by targeting human epidermal growth factor receptor 2 (HER2), a surface antigen expressed on tumor cells. ADCs comprise three main components: an antibody, a linker, and a cytotoxic payload. The antibody selectively binds to tumor cell surface antigens, facilitating targeted delivery of the cytotoxic drug, while linkers play a crucial role in ensuring stability and controlled release of the payload. Cleavable linkers release the drug within tumor cells, while non-cleavable linkers ensure stability during circulation. The cytotoxic payload exerts its antitumor effect by disrupting cellular pathways. HER2 is commonly overexpressed in UCs, making it a potential therapeutic target. Several ADCs targeting HER2 have been approved for cancer treatment, but their use in UC is still being tested. Numerous HER2 ADCs have demonstrated significant growth inhibition and induction of apoptosis in translational models of HER2-overexpressing bladder cancer. Ongoing clinical trials are assessing the efficacy and safety of ADCs targeting HER2 in UC, with the aim of determining tumor response and the potential of ADCs as a treatment option for UC patients. The development of effective therapies with improved response rates and long-term effectiveness is crucial for advanced and metastatic UC. ADCs targeting HER2 show promise in this regard and merit further investigation for UC treatment.
Collapse
Affiliation(s)
- Chia-Hsien Shih
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yu-Hua Lin
- Division of Urology, Department of Surgery, Cardinal Tien Hospital, New Taipei City, Taiwan
- Department of Chemistry, Fu Jen Catholic University, New Taipei City, Taiwan
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Hao-Lun Luo
- Department of Urology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Wen-Wei Sung
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Urology, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| |
Collapse
|
8
|
Mohanty SK, Lobo A, Mishra SK, Cheng L. Precision Medicine in Bladder Cancer: Present Challenges and Future Directions. J Pers Med 2023; 13:jpm13050756. [PMID: 37240925 DOI: 10.3390/jpm13050756] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Bladder cancer (BC) is characterized by significant histopathologic and molecular heterogeneity. The discovery of molecular pathways and knowledge of cellular mechanisms have grown exponentially and may allow for better disease classification, prognostication, and development of novel and more efficacious noninvasive detection and surveillance strategies, as well as selection of therapeutic targets, which can be used in BC, particularly in a neoadjuvant or adjuvant setting. This article outlines recent advances in the molecular pathology of BC with a better understanding and deeper focus on the development and deployment of promising biomarkers and therapeutic avenues that may soon make a transition into the domain of precision medicine and clinical management for patients with BC.
Collapse
Affiliation(s)
- Sambit K Mohanty
- Department of Pathology and Laboratory Medicine, Advanced Medical Research Institute and CORE Diagnostics, Gurgaon 122016, India
| | - Anandi Lobo
- Department of Pathology and Laboratory Medicine, Kapoor Center for Pathology and Urology, Raipur 490042, India
| | - Sourav K Mishra
- Department of Medical Oncology, All India Institute of Medical Sciences, Bhubaneswar 750017, India
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Brown University Warren Alpert Medical School, Lifespan Academic Medical Center, and the Legorreta Cancer Center at Brown University, 593 Eddy Street, APC 12-105, Providence, RI 02903, USA
| |
Collapse
|
9
|
Sanguedolce F, Zanelli M, Palicelli A, Bisagni A, Zizzo M, Ascani S, Pedicillo MC, Cormio A, Falagario UG, Carrieri G, Cormio L. HER2 Expression in Bladder Cancer: A Focused View on Its Diagnostic, Prognostic, and Predictive Role. Int J Mol Sci 2023; 24:ijms24043720. [PMID: 36835131 PMCID: PMC9962688 DOI: 10.3390/ijms24043720] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/04/2023] [Accepted: 02/07/2023] [Indexed: 02/15/2023] Open
Abstract
Bladder cancer (BC) is a heterogeneous disease from a molecular, morphological, and clinical standpoint. HER2 is a known oncogene involved in bladder carcinogenesis. Assessing HER2 overexpression as a result of its molecular changes in a routine pathology practice using immunohistochemistry might be a useful adjunct in several scenarios, namely (1) to correctly identify flat urothelial lesions and inverted urothelial lesions in the diagnostic setting; (2) to provide prognostic hints in both non-muscle invasive (NMI) and muscle invasive (MI) tumors, thus supplementing risk stratification tools, especially when evaluating higher-risk tumors such as those with variant morphology; (3) to improve antibody panels as a surrogate marker of BC molecular subtyping. Furthermore, the potential of HER2 as a therapeutic target has been only partly explored so far, in light of the ongoing development of novel target therapies.
Collapse
Affiliation(s)
- Francesca Sanguedolce
- Pathology Unit, Policlinico Riuniti, University of Foggia, 71122 Foggia, Italy
- Correspondence:
| | - Magda Zanelli
- Pathology Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Andrea Palicelli
- Pathology Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Alessandra Bisagni
- Pathology Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Maurizio Zizzo
- Surgical Oncology Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Stefano Ascani
- Pathology Unit, Azienda Ospedaliera Santa Maria di Terni, University of Perugia, 05100 Terni, Italy
| | | | - Angelo Cormio
- Urology Unit, Azienda Ospedaliero-Universitaria Ospedali Riuniti Di Ancona, Università Politecnica Delle Marche, 60126 Ancona, Italy
| | - Ugo Giovanni Falagario
- Department of Urology and Renal Transplantation, Policlinico Riuniti, University of Foggia, 71122 Foggia, Italy
| | - Giuseppe Carrieri
- Department of Urology and Renal Transplantation, Policlinico Riuniti, University of Foggia, 71122 Foggia, Italy
| | - Luigi Cormio
- Department of Urology and Renal Transplantation, Policlinico Riuniti, University of Foggia, 71122 Foggia, Italy
- Department of Urology, Bonomo Teaching Hospital, 76123 Andria, Italy
| |
Collapse
|
10
|
Gandhi J, Chen JF, Al-Ahmadie H. Urothelial Carcinoma: Divergent Differentiation and Morphologic Subtypes. Surg Pathol Clin 2022; 15:641-659. [PMID: 36344181 PMCID: PMC9756812 DOI: 10.1016/j.path.2022.07.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Urothelial carcinoma (UC) is known to encompass a wide spectrum of morphologic features and molecular alterations. Approximately 15% to 25% of invasive UC exhibits histomorphologic features in the form of "divergent differentiation" along other epithelial lineages, or different "subtypes" of urothelial or sarcomatoid differentiation. It is recommended that the percentage of divergent differentiation and or subtype(s) be reported whenever possible. Recent advances in molecular biology have led to a better understanding of the molecular underpinning of these morphologic variations. In this review, we highlight histologic characteristics of the divergent differentiation and subtypes recognized by the latest version of WHO classification, with updates on their molecular and clinical features.
Collapse
Affiliation(s)
- Jatin Gandhi
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, 1364 Clifton Rd, Atlanta, GA 30322, USA
| | - Jie-Fu Chen
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Hikmat Al-Ahmadie
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
11
|
Abstract
PURPOSE OF REVIEW This review will discuss micropapillary urothelial carcinoma with respect to biology, histopathologic characteristics, genetic and molecular features, diagnosis, clinical management, and future directions of research. RECENT FINDINGS Recent consensus opinion study showed only moderate interobserver reproducibility in the diagnostic criteria. The most reproducible criteria with the highest consensus were multiple nests in the same lacunar spaces. There are recent reports of high rates of intratumoral heterogeneity of ERBB2 amplification within tumor containing both micropapillary and classic urothelial components. Micropapillary urothelial carcinoma is a well-documented highly aggressive variant of urothelial carcinoma with proven worse outcomes. Accurate recognition and reporting of this pattern is critical for optimal management. Newer therapeutic strategies related to the molecular and genetic findings seen in MPUC remain to be explored further.
Collapse
|
12
|
Abstract
Urothelial carcinoma is characterized by the presence of a wide spectrum of histopathologic features and molecular alterations that contribute to its morphologic and genomic heterogeneity. It typically harbors high rates of somatic mutations with considerable genomic and transcriptional complexity and heterogeneity that is reflective of its varied histomorphologic and clinical features. This review provides an update on the recent advances in the molecular characterization and novel molecular taxonomy of urothelial carcinoma and variant histologies.
Collapse
Affiliation(s)
- Hikmat Al-Ahmadie
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NW 10065, USA.
| | - George J Netto
- Department of Pathology, University of Alabama at Birmingham, University of Alabama at Birmingham School of Medicine, WP Building, Suite P230, 619 19th Street South, Birmingham, AL 35249-7331, USA.
| |
Collapse
|
13
|
Sung YN, Kim SJ, Jun SY, Yoo C, Kim KP, Lee JH, Hwang DW, Hwang S, Lee SS, Hong SM. Expression of HER2 and Mismatch Repair Proteins in Surgically Resected Gallbladder Adenocarcinoma. Front Oncol 2021; 11:658564. [PMID: 34367955 PMCID: PMC8339709 DOI: 10.3389/fonc.2021.658564] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 06/29/2021] [Indexed: 12/30/2022] Open
Abstract
Background Gallbladder cancer (GBC) has a poor prognosis. Although complete surgical resection is the only successful approach for improving survival, additional therapeutic modalities are required for recurrent or surgically unresectable GBCs. Materials and Methods To determine the expression status of HER2 and the mismatch repair (MMR) proteins MLH1, MSH2, MSH6, and PMS2, immunohistochemical staining of MMR proteins and HER2 was carried out in 216 surgically resected GBCs. HER2 labeling was scored by adopting a scoring system for gastric carcinomas. Tissues scoring 0 to 2+ were defined as HER2 negative, whereas those scoring 3+ were regarded as HER2-positive. In addition, silver in situ hybridization and microsatellite instability (MSI) analysis were conducted to confirm HER2 amplification and MSI, respectively. Results Three of 216 GBCs (1.3%) showed MMR protein deficiency. All three observed MSI cases exhibited dual loss of MSH2 and MSH6 protein expression. However, no cases showed loss of either MLH1 or PMS2 expression. No association was observed between MMR protein deficiency and other clinicopathological factors. HER2 amplification was noted in 30 (13.9%) GBCs and associated with Crohn-like lymphoid reaction (P = 0.023). No survival difference was observed based on HER2 overexpression or HER2 amplification status. Conclusion MMR protein deficiency and HER2 overexpression were observed in a small subset (1.3% and 13.9%, respectively) of GBCs without simultaneous occurrence of deficient MMR protein expression and HER2 overexpression. The presence of Crohn-like lymphoid reaction may help identify cases with HER2 amplification, by using hematoxylin-stained slides. Although the proportion of MMR protein-deficient- and HER2-overexpressing GBCs was small, applying immunotherapy to MMR protein-deficient GBCs and herceptin to HER2-overexpressing GBCs may provide alternative treatment options for patients with GBC.
Collapse
Affiliation(s)
- You-Na Sung
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Sung Joo Kim
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Sun-Young Jun
- Department of Pathology, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Changhoon Yoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Kyu-Pyo Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jae Hoon Lee
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Dae Wook Hwang
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Shin Hwang
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Sang Soo Lee
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Seung-Mo Hong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| |
Collapse
|
14
|
Li Y, Sun L, Guo X, Mo N, Zhang J, Li C. Frontiers in Bladder Cancer Genomic Research. Front Oncol 2021; 11:670729. [PMID: 34094968 PMCID: PMC8173177 DOI: 10.3389/fonc.2021.670729] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 04/28/2021] [Indexed: 02/05/2023] Open
Abstract
Most of the etiology studies of bladder cancer focus on genetic changes, mainly including mutation and activation of oncogenes, mutation and inactivation of tumor suppressor genes, and rearrangement or heterozygous deletion of chromosomes. Moreover, bladder cancer is highly heterogeneous mainly due to abnormal changes in the genome and proteome of tumor cells. Surgery is the main treatment for bladder cancer, but because the recurrence rate is high after surgery and most of the muscle-invasive bladder cancer acquires distant metastasis. Therefore, there is a need to combine with chemotherapy to consolidate the treatment effect. However, there are differences in chemosensitivity among patients. In this article, we review the up-to-date genomic researches on bladder cancer occurrence, development, metastasis, and chemosensitivity in patients, in order to provide some theoretical support for the diagnosis and treatment strategy for bladder cancer.
Collapse
Affiliation(s)
- Yi Li
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Lihui Sun
- Core Facility for Protein Research, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xiangyang Guo
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Na Mo
- Department of Pathology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Jinku Zhang
- Department of Pathology, First Central Hospital of Baoding, Baoding, China.,Key Laboratory of Molecular Pathology and Early Diagnosis of Tumor in Hebei Province, First Central Hospital of Baoding, Baoding, China
| | - Chong Li
- Core Facility for Protein Research, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,Key Laboratory of Molecular Pathology and Early Diagnosis of Tumor in Hebei Province, First Central Hospital of Baoding, Baoding, China.,Department of Immunology, Beijing Jianlan Institute of Medicine, Beijing, China.,Department of Immunology, Beijing Zhongke Jianlan Biotechnology Co., Ltd., Beijing, China
| |
Collapse
|
15
|
Moradi Tabriz H, Nazar E, Ahmadi SA, Azimi E, Majidi F. Survivin and Her2 Expressions in Different Grades of Urothelial Neoplasms of Urinary Bladder. IRANIAN JOURNAL OF PATHOLOGY 2020; 16:154-161. [PMID: 33936226 PMCID: PMC8085283 DOI: 10.30699/ijp.2020.130859.2447] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 08/31/2020] [Indexed: 02/03/2023]
Abstract
Background & Objective: Urothelial neoplasm (UN) of bladder is a potentially lethal malignancy, particularly in locally advanced or metastatic cases. Development of molecular markers such as HER2 and Survivin may provide useful information on diagnosis and prognosis in UN of bladder. Methods: We studied the immunohistochemical (IHC) expression of HER2 and Survivin in 84 radical/partial cystectomy and transurethral resection (TUR) specimens with different histologic grades and stages. All samples were obtained from Pathology Department of Sina Hospital in Tehran, Iran from 2014 to 2018. Results: From the total number of 84 UN samples, 10 cases (11.9%) showed papillary neoplasm of low malignant potential, 30 cases (35.7%) presented with low-grade papillary urothelial neoplasm, and 44 cases (52.4%) diagnosed as high-grade papillary urothelial neoplasm. HER2 and Survivin expressions were seen in 44 (52.4%) (P=0.610) and 9 (10.7%) patients (P=0.046), respectively. Survivin expression showed a mild increase in high grade UN. Conclusion: Our findings suggest that the IHC expression of Survivin and HER2 are not well associated with histological grades of urothelial neoplasms of bladder. This may be partly due to relatively small sample size and other factors such as patient characteristics or antibody specifications.
Collapse
Affiliation(s)
- Hedieh Moradi Tabriz
- Department of Pathology, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Nazar
- Department of Pathology, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Ali Ahmadi
- Department of Pathology, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Esmaeil Azimi
- Department of Pathology, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Fazeleh Majidi
- Research and Development Center Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Franceschini T, Capizzi E, Massari F, Schiavina R, Fiorentino M, Giunchi F. Immunohistochemical over-expression of HER2 does not always match with gene amplification in invasive bladder cancer. Pathol Res Pract 2020; 216:153012. [PMID: 32703487 DOI: 10.1016/j.prp.2020.153012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 05/06/2020] [Accepted: 05/10/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND HER2 is a potential target of therapy in urothelial cancer (UC). Pathological case stratification according to HER2 gene amplification or HER2 protein overexpression was critical for patients' selection in previous unsuccessful clinical trial with HER2 targeting agents. STUDY DESIGN We evaluated the HER2 overexpression by immunohistochemistry (IHC) together with the amplification of the HER2 gene with chromogenic(CISH) and fluorescent (FISH) in situ hybridization in a cohort of 61 patients covering the whole spectrum of bladder UC variants, using a tissue microarray (TMA) approach. RESULTS IHC was available in all the 61 cases while ISH in 37 and FISH in 42. At IHC, 2/61 cases (3%) were scored 3+; 2 (3%) scored 2+; 2 (3%) scored 1+; the remaining 55 (91%) scored 0. At CISH analysis 10/37 cases (27%) were amplified, 6 cases with HER2 amplification showed positive HER2 IHC (3+, 2+, 1+). Seven cases with IHC score 0 were amplified at CISH. FISH analysis revealed an amplification in 5/42 cases (12%). The total number of HER2amplified cases was different between chromogenic and fluorescent ISH with 5 cases amplified using FISH compared to 10 with CISH. CONCLUSIONS In clinical trials with HER2 targeting agents the candidate patients should be investigated not only by IHC but also by ISH, independently of the IHC results. Since also usual type UC can overexpress HER2 we recommend to extend the patients' selection to all the histotypes of bladder cancer other than the micropapillary type.
Collapse
Affiliation(s)
- Tania Franceschini
- Department of Pathology, S.Orsola and Maggiore Hospital, and Department of Specialistic Diagnostic and Experimental Medicine, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Elisa Capizzi
- Department of Pathology, S.Orsola and Maggiore Hospital, and Department of Specialistic Diagnostic and Experimental Medicine, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Francesco Massari
- Department of Oncology, S.Orsola Hospital, and Department of Specialistic Diagnostic and Experimental Medicine, University of Bologna, Bologna, Italy
| | - Riccardo Schiavina
- Department of Urology, S.Orsola Hospital, and Department of Specialistic Diagnostic and Experimental Medicine, University of Bologna, Bologna, Italy
| | - Michelangelo Fiorentino
- Department of Pathology, S.Orsola and Maggiore Hospital, and Department of Specialistic Diagnostic and Experimental Medicine, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy.
| | - Francesca Giunchi
- Department of Pathology, S.Orsola and Maggiore Hospital, and Department of Specialistic Diagnostic and Experimental Medicine, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| |
Collapse
|
17
|
Abstract
Urothelial carcinoma (UC) is the most common malignant neoplasm of the bladder that encompasses a wide spectrum of histopathologic features and various molecular alterations and subtypes, responsible for its significant morphologic and genomic heterogeneity. Morphologically, in addition to classic UC (not otherwise specified), many well documented variant histologies are a common finding in invasive UC, and include squamous, glandular, micropapillary, sarcomatoid, small cell/neuroendocrine, clear cell, lymphoepithelioma-like, and plasmacytoid types, among others. This review provides an update on the recent advances in the molecular characterization and novel molecular taxonomy of UC and variant histologies.
Collapse
|
18
|
McGregor BA, Sonpavde G. Enfortumab Vedotin, a fully human monoclonal antibody against Nectin 4 conjugated to monomethyl auristatin E for metastatic urothelial Carcinoma. Expert Opin Investig Drugs 2019; 28:821-826. [PMID: 31526130 DOI: 10.1080/13543784.2019.1667332] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/10/2019] [Indexed: 12/27/2022]
Abstract
Introduction: The conventional management of most patients with metastatic urothelial carcinoma (UC) is platinum-based chemotherapy followed by immunotherapy. Erdafitinib is an option in post-platinum patients with activating mutations in fibroblast growth factor receptor (FGFR)-3 and -2. Salvage therapy with taxanes or vinflunine has demonstrated minimal efficacy. Enfortumab Vedotin (EV), a monoclonal antibody-drug conjugate (ADC) targeting nectin-4 is under investigation in patients with advanced UC. Areas covered: This review describes the epidemiology and unmet needs of patients with metastatic UC and is focused specifically on heavily treated patients. We explore the rationale for targeting nectin 4 and the clinical development of EV; efficacy and safety data from the completed phase I and II studies are examined. Ongoing trials to definitively assess clinical outcomes in comparison to current therapy and trials exploring EV in combination are also highlighted. Expert opinion: There is an unmet need for new therapies in most patients with advanced UC and who progress after platinum and immunotherapy. EV has shown promising efficacy and safety in this population in phase 1 and 2 trials including those with poor prognostic factors such as liver metastases. Ongoing trials exploring this agent in combination will continue to advance the treatment of UC.
Collapse
Affiliation(s)
- Bradley A McGregor
- Lank Center for Genitourinary Oncology, Dana Farber Cancer Institute , Boston , MA , USA
- Department of Medicine, Harvard Medical School , Boston , MA , USA
| | - Guru Sonpavde
- Lank Center for Genitourinary Oncology, Dana Farber Cancer Institute , Boston , MA , USA
- Department of Medicine, Harvard Medical School , Boston , MA , USA
| |
Collapse
|
19
|
Genitsch V, Kollár A, Vandekerkhove G, Blarer J, Furrer M, Annala M, Herberts C, Pycha A, de Jong JJ, Liu Y, Krentel F, Davicioni E, Gibb EA, Kruithof-de Julio M, Wyatt AW, Seiler R. Morphologic and genomic characterization of urothelial to sarcomatoid transition in muscle-invasive bladder cancer. Urol Oncol 2019; 37:826-836. [PMID: 31585777 DOI: 10.1016/j.urolonc.2019.09.025] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 06/22/2019] [Accepted: 06/23/2019] [Indexed: 01/05/2023]
Abstract
INTRODUCTION The sarcomatoid morphology of muscle-invasive bladder cancer (MIBC) is associated with unfavorable prognosis. However, the genomic, transcriptomic, and proteomic relationship between conventional urothelial and synchronous sarcomatoid morphology is poorly defined. METHODS We compiled a cohort of 21 MIBC patients with components of conventional urothelial and adjacent sarcomatoid morphology within the same tumor focus. We performed comprehensive pathologic and immunohistochemical characterization and in 4 selected cases, subjected both morphologic components to targeted DNA sequencing and whole transcriptome analysis. RESULTS Synchronous sarcomatoid and urothelial morphology from the same MIBC foci shared truncal somatic mutations, indicating a common ancestral clone. However, additional mutations or copy number alterations restricted to the either component suggested divergent evolution at the genomic level. This was confirmed at the transcriptome level since while the urothelial component exhibited a basal-like subtype (TCGA2014: cluster III, LundTax: basal/squamous-like), the sarcomatoid morphology was predominantly cluster IV (claudin-low). Protein expression was consistent with a basal-like phenotype in both morphologies in 18/21 of cases. However, most cases had evidence of active epithelial-to-mesenchymal transition (E-Cad ↓ and Zeb1 or TWIST1 ↑) from urothelial toward the sarcomatoid morphology. Drug response signatures nominated different targets for each morphology and proposed agents under clinical investigation in liposarcoma or other sarcoma. PD-L1 expression was higher in the sarcomatoid than the urothelial component. CONCLUSIONS Conventional urothelial and adjacent sarcomatoid morphologies of MIBC arise from the same common ancestor and share a basal-like phenotype. However, divergence between the morphologies at the genome, transcriptome, and proteome level suggests differential sensitivity to therapy.
Collapse
Affiliation(s)
- Vera Genitsch
- Institute of Pathology, University of Bern, Switzerland
| | - Attila Kollár
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Gillian Vandekerkhove
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Jennifer Blarer
- Department of Urology, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Marc Furrer
- Department of Urology, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Matti Annala
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, Canada; Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Cameron Herberts
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Armin Pycha
- Department of Urology, Central Hospital of Bolzano, Bolzano, Italy
| | - Joep J de Jong
- Department of Urology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Yang Liu
- GenomeDx Inc., Vancouver, Canada
| | - Friedemann Krentel
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | | | | | | | - Alexander W Wyatt
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Roland Seiler
- Department of Urology, Inselspital, Bern University Hospital, University of Bern, Switzerland.
| |
Collapse
|
20
|
Sailer V, Eng KW, Zhang T, Bareja R, Pisapia DJ, Sigaras A, Bhinder B, Romanel A, Wilkes D, Sticca E, Cyrta J, Rao R, Sahota S, Pauli C, Beg S, Motanagh S, Kossai M, Fontugne J, Puca L, Rennert H, Xiang JZ, Greco N, Kim R, MacDonald TY, McNary T, Blattner-Johnson M, Schiffman MH, Faltas BM, Greenfield JP, Rickman D, Andreopoulou E, Holcomb K, Vahdat LT, Scherr DS, van Besien K, Barbieri CE, Robinson BD, Fine HA, Ocean AJ, Molina A, Shah MA, Nanus DM, Pan Q, Demichelis F, Tagawa ST, Song W, Mosquera JM, Sboner A, Rubin MA, Elemento O, Beltran H. Integrative Molecular Analysis of Patients With Advanced and Metastatic Cancer. JCO Precis Oncol 2019; 3:PO.19.00047. [PMID: 31592503 PMCID: PMC6778956 DOI: 10.1200/po.19.00047] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
PURPOSE We developed a precision medicine program for patients with advanced cancer using integrative whole-exome sequencing and transcriptome analysis. PATIENTS AND METHODS Five hundred fifteen patients with locally advanced/metastatic solid tumors were prospectively enrolled, and paired tumor/normal sequencing was performed. Seven hundred fifty-nine tumors from 515 patients were evaluated. RESULTS Most frequent tumor types were prostate (19.4%), brain (16.5%), bladder (15.4%), and kidney cancer (9.2%). Most frequently altered genes were TP53 (33%), CDKN2A (11%), APC (10%), KTM2D (8%), PTEN (8%), and BRCA2 (8%). Pathogenic germline alterations were present in 10.7% of patients, most frequently CHEK2 (1.9%), BRCA1 (1.5%), BRCA2 (1.5%), and MSH6 (1.4%). Novel gene fusions were identified, including a RBM47-CDK12 fusion in a metastatic prostate cancer sample. The rate of clinically relevant alterations was 39% by whole-exome sequencing, which was improved by 16% by adding RNA sequencing. In patients with more than one sequenced tumor sample (n = 146), 84.62% of actionable mutations were concordant. CONCLUSION Integrative analysis may uncover informative alterations for an advanced pan-cancer patient population. These alterations are consistent in spatially and temporally heterogeneous samples.
Collapse
Affiliation(s)
| | | | - Tuo Zhang
- Weill Cornell Medicine, New York, NY
| | | | | | | | | | | | | | | | | | - Rema Rao
- Weill Cornell Medicine, New York, NY
| | | | | | | | | | | | | | | | | | | | | | - Rob Kim
- Weill Cornell Medicine, New York, NY
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Qiulu Pan
- Weill Cornell Medicine, New York, NY
| | | | | | - Wei Song
- Weill Cornell Medicine, New York, NY
| | | | | | | | | | - Himisha Beltran
- Weill Cornell Medicine, New York, NY,Himisha Beltran, MD, Weill Cornell Medicine, 413 E. 69th Street, New York, NY 10021; e-mail:
| |
Collapse
|
21
|
Genitsch V, Kollár A, Vandekerkhove G, Blarer J, Furrer M, Annala M, Herberts C, Pycha A, de Jong JJ, Liu Y, Krentel F, Davicioni E, Gibb EA, Kruithof-de Julio M, Wyatt AW, Seiler R. Morphologic and genomic characterization of urothelial to sarcomatoid transition in muscle-invasive bladder cancer. Urol Oncol 2019; 37:573.e19-573.e29. [PMID: 31358384 DOI: 10.1016/j.urolonc.2019.06.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 06/22/2019] [Accepted: 06/23/2019] [Indexed: 10/26/2022]
Abstract
INTRODUCTION The sarcomatoid morphology of muscle-invasive bladder cancer (MIBC) is associated with unfavorable prognosis. However, the genomic, transcriptomic, and proteomic relationship between conventional urothelial and synchronous sarcomatoid morphology is poorly defined. METHODS We compiled a cohort of 21 MIBC patients with components of conventional urothelial and adjacent sarcomatoid morphology within the same tumor focus. We performed comprehensive pathologic and immunohistochemical characterization and in 4 selected cases, subjected both morphologic components to targeted DNA sequencing and whole transcriptome analysis. RESULTS Synchronous sarcomatoid and urothelial morphology from the same MIBC foci shared truncal somatic mutations, indicating a common ancestral clone. However, additional mutations or copy number alterations restricted to the either component suggested divergent evolution at the genomic level. This was confirmed at the transcriptome level since while the urothelial component exhibited a basal-like subtype (TCGA2014: cluster III, LundTax: basal/squamous-like), the sarcomatoid morphology was predominantly cluster IV (claudin-low). Protein expression was consistent with a basal-like phenotype in both morphologies in 18/21 of cases. However, most cases had evidence of active epithelial-to-mesenchymal transition (E-Cad ↓ and Zeb1 or TWIST1 ↑) from urothelial toward the sarcomatoid morphology. Drug response signatures nominated different targets for each morphology and proposed agents under clinical investigation in liposarcoma or other sarcoma. PD-L1 expression was higher in the sarcomatoid than the urothelial component. CONCLUSIONS Conventional urothelial and adjacent sarcomatoid morphologies of MIBC arise from the same common ancestor and share a basal-like phenotype. However, divergence between the morphologies at the genome, transcriptome, and proteome level suggests differential sensitivity to therapy.
Collapse
Affiliation(s)
- Vera Genitsch
- Institute of Pathology, University of Bern, Switzerland
| | - Attila Kollár
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Gillian Vandekerkhove
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Jennifer Blarer
- Department of Urology, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Marc Furrer
- Department of Urology, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Matti Annala
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, Canada; Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Cameron Herberts
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Armin Pycha
- Department of Urology, Central Hospital of Bolzano, Bolzano, Italy
| | - Joep J de Jong
- Department of Urology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Yang Liu
- GenomeDx Inc., Vancouver, Canada
| | - Friedemann Krentel
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | | | | | | | - Alexander W Wyatt
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Roland Seiler
- Department of Urology, Inselspital, Bern University Hospital, University of Bern, Switzerland.
| |
Collapse
|
22
|
Sanguedolce F, Russo D, Mancini V, Selvaggio O, Calo B, Carrieri G, Cormio L. Prognostic and therapeutic role of HER2 expression in micropapillary carcinoma of the bladder. Mol Clin Oncol 2019; 10:205-213. [PMID: 30680196 PMCID: PMC6327213 DOI: 10.3892/mco.2018.1786] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 11/22/2018] [Indexed: 12/18/2022] Open
Abstract
Micropapillary carcinoma of the bladder (MPBC) is a variant type of infiltrating urothelial carcinoma, which portends a poor biological behavior in terms of disease stage at first diagnosis and clinical outcome; its peculiar morphology raises issues concerning the ability of tumor detection by imaging techniques and proper biopsy procedure, and the appropriate treatment for non-muscle infiltrating and muscle-infiltrating MPBC remains a matter of debate. On the basis of its established prognostic and therapeutic role in breast and gastro-esophageal cancer in the first instance, the human epidermal growth factor receptor-2 (HER2) has been investigated in selected case series of MPBC over the last 10 years. The aim of the present review was to summarize the existing evidence on HER2 status in MPBC, and to discuss its present and future utility in risk assessment and treatment choice of this uncommon, yet aggressive, disease.
Collapse
Affiliation(s)
| | - Davide Russo
- Department of Pathology, University Hospital, I-71121 Foggia, Italy
| | - Vito Mancini
- Department of Urology and Renal Transplantation, University Hospital, I-71121 Foggia, Italy
| | - Oscar Selvaggio
- Department of Urology and Renal Transplantation, University Hospital, I-71121 Foggia, Italy
| | - Beppe Calo
- Department of Urology and Renal Transplantation, University Hospital, I-71121 Foggia, Italy
| | - Giuseppe Carrieri
- Department of Urology and Renal Transplantation, University Hospital, I-71121 Foggia, Italy
| | - Luigi Cormio
- Department of Urology and Renal Transplantation, University Hospital, I-71121 Foggia, Italy
| |
Collapse
|
23
|
Updates on the Genetics and Molecular Subtypes of Urothelial Carcinoma and Select Variants. Surg Pathol Clin 2018; 11:713-723. [PMID: 30447837 DOI: 10.1016/j.path.2018.07.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Urothelial carcinoma is a morphologically and genomically heterogeneous disease that exhibits a wide spectrum of morphologic features and molecular alterations and subtypes. Classic urothelial carcinoma (not otherwise specified) is the most common tumor type that develops in the urinary bladder but many, well-documented, variant histologies are commonly encountered in approximately one-third of invasive urothelial carcinoma, including squamous, glandular, micropapillary, sarcomatoid, small cell/neuroendocrine, clear cell, lymphoepithelioma-like, and plasmacytoid types, among others. In this review, we provide an update on the molecular advances in urothelial carcinoma and some of its variant histologies.
Collapse
|
24
|
Zinnall U, Weyerer V, Compérat E, Camparo P, Gaisa NT, Knuechel-Clarke R, Perren A, Lugli A, Toma M, Baretton G, Kristiansen G, Wirtz RM, Cheng L, Wullich B, Stoehr R, Hartmann A, Bertz S. Micropapillary urothelial carcinoma: evaluation of HER2 status and immunohistochemical characterization of the molecular subtype. Hum Pathol 2018; 80:55-64. [DOI: 10.1016/j.humpath.2018.05.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/11/2018] [Accepted: 05/15/2018] [Indexed: 11/28/2022]
|
25
|
Reappraisal of HER2 status in the spectrum of advanced urothelial carcinoma: a need of guidelines for treatment eligibility. Mod Pathol 2018; 31:1270-1281. [PMID: 29467478 DOI: 10.1038/s41379-018-0023-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 12/29/2017] [Accepted: 12/30/2017] [Indexed: 01/05/2023]
Abstract
Although human epidermal growth factor receptor 2 (HER2) may represent a therapeutic target, its evaluation in urothelial carcinoma of the bladder does not rely on a standardized scoring system by immunohistochemistry or fluorescent in situ hybridization (FISH), as reflected by various methodology in the literature and clinical trials. Our aim was to improve and standardize HER2 amplification detection in bladder cancer. We assessed immunohistochemical criteria derived from 2013 American Society of Clinical Oncology (ASCO)/College of American Pathologists (CAPs) guidelines for breast cancer and investigated intratumoral heterogeneity in a retrospective multicentric cohort of 188 patients with locally advanced urothelial carcinoma of the bladder. Immunohistochemistry was performed on 178 primary tumors and 126 lymph node metastases, eligible cases (moderate/strong, complete/incomplete membrane staining) were assessed by FISH. HER2 overexpression was more frequent with 2013 ASCO/CAP than 2007 ASCO/CAP guidelines (p < 0.0001). The rate of positive HER2 FISH was similar between primary tumor and lymph node metastases (8%). Among positive FISH cases, 48% were associated with moderate/strong incomplete membrane staining that were not scored eligible for FISH by 2007 ASCO/CAP criteria. Among 3+ immunohistochemistry score cases, 67% were associated with HER2-positive FISH. Concordance between primary tumors and matched lymph node metastases was moderate for immunohistochemistry (κ = 0.54 (CI 95%, 0.41-0.67)) and FISH (κ = 0.50 (CI 95%, 0.20-0.79)). HER2-positive FISH was more frequent in micropapillary carcinomas (12%) and carcinoma with squamous differentiation (11%) than in pure conventional carcinoma (6%). Intratumoral heterogeneity for HER2 immunohistochemistry was observed in 7% primary tumor and 6% lymph node metastases; 24% positive HER2 FISH presented intratumoral heterogeneity. Our study suggests that HER2 evaluation should include an immunohistochemistry screening step with eligibility for FISH including incomplete/complete and moderate/strong membrane staining. Spatial or temporal intratumoral heterogeneity prompts to perform evaluation on both tumor and lymph node, and for each histological variant observed.
Collapse
|
26
|
Vlachostergios PJ, Jakubowski CD, Niaz MJ, Lee A, Thomas C, Hackett AL, Patel P, Rashid N, Tagawa ST. Antibody-Drug Conjugates in Bladder Cancer. Bladder Cancer 2018; 4:247-259. [PMID: 30112436 PMCID: PMC6087439 DOI: 10.3233/blc-180169] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Urothelial carcinoma (UC) is characterized by expression of a plethora of cell surface antigens, thus offering opportunities for specific therapeutic targeting with use of antibody-drug conjugates (ADCs). ADCs are structured from two major constituents, a monoclonal antibody (mAb) against a specific target and a cytotoxic drug connected via a linker molecule. Several ADCs are developed against different UC surface markers, but the ones at most advanced stages of development include sacituzumab govitecan (IMMU-132), enfortumab vedotin (ASG-22CE/ASG-22ME), ASG-15ME for advanced UC, and oportuzumab monatox (VB4-845) for early UC. Several new targets are identified and utilized for novel or existing ADC testing. The most promising ones include human epidermal growth factor receptor 2 (HER2) and members of the fibroblast growth factor receptor axis (FGF/FGFR). Positive preclinical and early clinical results are reported in many cases, thus the next step involves further improving efficacy and reducing toxicity as well as testing combination strategies with approved agents.
Collapse
Affiliation(s)
| | | | - Muhammad J. Niaz
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Aileen Lee
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Charlene Thomas
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Amy L. Hackett
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Priyanka Patel
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Naureen Rashid
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Scott T. Tagawa
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Department of Urology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
27
|
Behzatoğlu K, Yörükoğlu K, Demir H, Bal N. Human Epidermal Growth Factor Receptor 2 Overexpression in Micropapillary and Other Variants of Urothelial Carcinoma. Eur Urol Focus 2018; 4:399-404. [DOI: 10.1016/j.euf.2016.06.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 05/29/2016] [Accepted: 06/11/2016] [Indexed: 01/06/2023]
|
28
|
Isharwal S, Huang H, Nanjangud G, Audenet F, Chen YB, Gopalan A, Fine SW, Tickoo SK, Lee BH, Iyer G, Chadalavada K, Rosenberg JE, Bajorin DF, Herr HW, Donat SM, Dalbagni G, Bochner BH, Solit DB, Reuter VE, Al-Ahmadie HA. Intratumoral heterogeneity of ERBB2 amplification and HER2 expression in micropapillary urothelial carcinoma. Hum Pathol 2018; 77:63-69. [PMID: 29601842 DOI: 10.1016/j.humpath.2018.03.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 03/06/2018] [Accepted: 03/19/2018] [Indexed: 01/12/2023]
Abstract
Micropapillary urothelial carcinoma (MPUC) is a rare but an aggressive variant of urothelial carcinoma. MPUC has been shown to commonly exhibit ERBB2 amplification and HER2 protein overexpression, but the frequency and distribution of these findings within micropapillary (MP) and not otherwise specified (NOS) components of tumors with mixed histology have not been addressed. Therefore, we evaluated ERBB2 amplification and HER2 expression in 43 MPUC cases by fluorescence in situ hybridization (FISH) and immunohistochemistry (IHC). Of the 35 tumors containing both MP and NOS components, ERBB2 amplification was present in both the MP and NOS components of 12 tumors (34.3%), in only the MP component of 11 tumors (31.4%), and exclusively in the NOS component of 4 tumors (11.4%). HER2 protein overexpression was significantly more commonly present in the MP component compared to the NOS component within the same tumor (68.6% versus 34.3%, P = .012). Overall, there was a moderately positive correlation between HER2 protein expression and ERBB2 amplification in both MP (ρ = 0.59, P < .001) and NOS (ρ = 0.70, P < .001) components. All MP/NOS areas with IHC score 3+ and none of MP/NOS areas with IHC score 0 were associated with ERBB2 amplification. We conclude that ERBB2 amplification and HER2 overexpression are preferentially but not exclusively identified in the MP component compared to the NOS component within the same tumor. Our findings identify the presence of intratumoral heterogeneity of ERBB2 amplification and HER2 expression in MPUC and provide grounds for further investigation into the mechanisms underlying the development of MPUC.
Collapse
Affiliation(s)
- Sumit Isharwal
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Hongying Huang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Gouri Nanjangud
- Molecular Cytogenetics Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - François Audenet
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Ying-Bei Chen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Anuradha Gopalan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Samson W Fine
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Satish K Tickoo
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Byron H Lee
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Gopa Iyer
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Kalyani Chadalavada
- Molecular Cytogenetics Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Jonathan E Rosenberg
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Dean F Bajorin
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Harry W Herr
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - S Machele Donat
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Guido Dalbagni
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Bernard H Bochner
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - David B Solit
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Victor E Reuter
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Hikmat A Al-Ahmadie
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065.
| |
Collapse
|
29
|
Zong H, Han L, Ding K, Wang J, Sun T, Zhang X, Cagliero C, Jiang H, Xie Y, Xu J, Zhang B, Zhu J. Producing defucosylated antibodies with enhanced in vitro antibody-dependent cellular cytotoxicity via FUT8 knockout CHO-S cells. Eng Life Sci 2017; 17:801-808. [PMID: 32624826 DOI: 10.1002/elsc.201600255] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 02/15/2017] [Accepted: 02/21/2017] [Indexed: 12/21/2022] Open
Abstract
To engineer a host cell line that produces defucosylated mAbs with superior antibody-dependent cellular cytotoxicity, we disrupted α-1, 6 fucosyltransferase (FUT8) gene in CHO-S (CHO is Chinese hamster ovary) cells by clustered regularly interspaced short palindromic repeats-CRISPR associated nuclease 9. The gene knockout cell line was evaluated for growth, stability, and product quality. The growth profile of FUT8 gene knockout CHO-S (FUT8 -/-) cells was comparable with wild type CHO-S cells. FUT8 catalyzes the transfer of a fucose residue from GDP-fucose to N-glycans residue. Defucosylated IgG1 antibodies produced by FUT8 -/- cells showed increased binding affinities to human FcγRIIIa and higher activities in mediating antibody-dependent cellular cytotoxicity, comparing with conventional fucosylated IgG1. Our results demonstrated the potential of using the clustered regularly interspaced short palindromic repeats-CRISPR associated nuclease 9 technology in cell line engineering for biopharmaceutical industrial applications.
Collapse
Affiliation(s)
- Huifang Zong
- Engineering Research Center of Cell & Therapeutic Antibody Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University Shanghai People's Republic of China
| | - Lei Han
- Engineering Research Center of Cell & Therapeutic Antibody Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University Shanghai People's Republic of China
| | - Kai Ding
- Engineering Research Center of Cell & Therapeutic Antibody Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University Shanghai People's Republic of China
| | - Jiaxian Wang
- Engineering Research Center of Cell & Therapeutic Antibody Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University Shanghai People's Republic of China
| | - Tao Sun
- Engineering Research Center of Cell & Therapeutic Antibody Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University Shanghai People's Republic of China
| | - Xinyu Zhang
- Engineering Research Center of Cell & Therapeutic Antibody Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University Shanghai People's Republic of China
| | | | | | | | - Jianrong Xu
- School of Medicine Shanghai Jiao Tong University Shanghai People's Republic of China
| | - Baohong Zhang
- Engineering Research Center of Cell & Therapeutic Antibody Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University Shanghai People's Republic of China
| | - Jianwei Zhu
- Engineering Research Center of Cell & Therapeutic Antibody Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University Shanghai People's Republic of China
- Jecho Laboratories Frederick MD USA
| |
Collapse
|
30
|
Abstract
Administration of neoadjuvant chemotherapy preceding radical cystectomy in patients with bladder cancer remains a matter of debate. Results of prospective, randomized studies have demonstrated an overall absolute survival benefit of 5% at 5 years, provided cisplatin-based combination regimens are used. Owing to the perception of a modest survival benefit, the medical community has been slow to adopt the use of neoadjuvant chemotherapy. Other reasons for the underuse of neoadjuvant chemotherapy range from patient ineligibility to fear of delaying potentially curative surgery in nonresponders. Instead, several institutions have adopted an individualized, risk-adapted approach, in which the decision to administer chemotherapy is based on clinical stage and patient comorbidity profile. The development of new cytotoxic and targeted therapies, in particular immune checkpoint inhibitors, warrants well-designed prospective studies to test their efficacy alone or in combination in the neoadjuvant setting. Moving forward, genomic characterization of muscle-invasive bladder cancer could offer information that aids clinicians in selecting the appropriate chemotherapy regimen. Following neoadjuvant therapy, every effort should be made to ensure optimal surgery, as surgical margins and the number of removed lymph nodes are prognostic factors; thus, radical cystectomy and a meticulous extended pelvic lymph node dissection should be performed by expert surgeons.
Collapse
|
31
|
Kiss B, Wyatt AW, Douglas J, Skuginna V, Mo F, Anderson S, Rotzer D, Fleischmann A, Genitsch V, Hayashi T, Neuenschwander M, Buerki C, Davicioni E, Collins C, Thalmann GN, Black PC, Seiler R. Her2 alterations in muscle-invasive bladder cancer: Patient selection beyond protein expression for targeted therapy. Sci Rep 2017; 7:42713. [PMID: 28205537 PMCID: PMC5311866 DOI: 10.1038/srep42713] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 01/12/2017] [Indexed: 12/22/2022] Open
Abstract
Although the introduction of novel targeted agents has improved patient outcomes in several human cancers, no such advance has been achieved in muscle-invasive bladder cancer (MIBC). However, recent sequencing efforts have begun to dissect the complex genomic landscape of MIBC, revealing distinct molecular subtypes and offering hope for implementation of targeted therapies. Her2 (ERBB2) is one of the most established therapeutic targets in breast and gastric cancer but agents targeting Her2 have not yet demonstrated anti-tumor activity in MIBC. Through an integrated analysis of 127 patients from three centers, we identified alterations of Her2 at the DNA, RNA and protein level, and demonstrate that Her2 relevance as a tumor driver likely may vary even within ERBB2 amplified cases. Importantly, tumors with a luminal molecular subtype have a significantly higher rate of Her2 alterations than those of the basal subtype, suggesting that Her2 activity is also associated with subtype status. Although some of our findings present rare events in bladder cancer, our study suggests that comprehensively assessing Her2 status in the context of tumor molecular subtype may help select MIBC patients most likely to respond to Her2 targeted therapy.
Collapse
Affiliation(s)
- Bernhard Kiss
- Department of Urology, University of Bern, Bern, Switzerland
| | - Alexander W Wyatt
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - James Douglas
- Department of Urology, University Hospital of Southampton, Hampshire SO16 6YD, UK
| | | | - Fan Mo
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Shawn Anderson
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Diana Rotzer
- Department of Urology, University of Bern, Bern, Switzerland
| | | | - Vera Genitsch
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Tetsutaro Hayashi
- Department of Urology, Institute of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
| | | | | | - Elai Davicioni
- GenomeDx Biosciences, Inc., Vancouver, British Columbia, Canada
| | - Colin Collins
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | | | - Peter C Black
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Roland Seiler
- Department of Urology, University of Bern, Bern, Switzerland.,Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| |
Collapse
|
32
|
Bertz S, Hartmann A, Knüchel-Clarke R, Gaisa NT. [Specific types of bladder cancer]. DER PATHOLOGE 2017; 37:40-51. [PMID: 26782034 DOI: 10.1007/s00292-015-0129-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Bladder cancer shows rare variants and special subtypes with diverse prognostic importance and therefore may necessitate different therapeutic approaches. For pathologists it is important to histologically diagnose and specify such variants. Nested variants of urothelial carcinoma with inconspicuous, well-formed tumor cell nests present with an aggressive course. The plasmacytoid variant, which morphologically resembles plasma cells is associated with a shorter survival time and a high frequency of peritoneal metastasis. Micropapillary urothelial carcinoma with small papillary tumor cell islands within artificial tissue retraction spaces and frequent lymphovascular invasion also has a poor prognosis. Other important rare differential variants listed in the World Health Organization (WHO) classification are microcystic, lymphoepithelioma-like, sarcomatoid, giant cell and undifferentiated urothelial carcinomas. Additionally, there are three special types of bladder cancer: squamous cell carcinoma, adenocarcinoma and small cell neuroendocrine carcinoma of the bladder. These tumors are characterized by pure squamous cell or glandular differentiation and are sometimes less responsive to adjuvant (chemo)therapy. Small cell carcinoma of the bladder mimics the neuroendocrine features of its pulmonary counterpart, shows an aggressive course but is sensitive to (neo-)adjuvant chemotherapy. The morphology and histology of the most important variants and special types are discussed in this review.
Collapse
Affiliation(s)
- S Bertz
- Institut für Pathologie, Universitätsklinikum Erlangen, Krankenhausstrasse 8-10, 91054, Erlangen, Deutschland.
| | - A Hartmann
- Institut für Pathologie, Universitätsklinikum Erlangen, Krankenhausstrasse 8-10, 91054, Erlangen, Deutschland
| | - R Knüchel-Clarke
- Institut für Pathologie, Uniklinikum RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Deutschland
| | - N T Gaisa
- Institut für Pathologie, Uniklinikum RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Deutschland.
| |
Collapse
|
33
|
Shawky Holah N, Abd El-Halim Kandil M, Abdel Razek E. A study of the prognostic and predictive role of HER-2 expression in bladder urothelial carcinoma. EGYPTIAN JOURNAL OF PATHOLOGY 2016; 36:241-250. [DOI: 10.1097/01.xej.0000508560.88111.51] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
34
|
Micropapillary morphology is an indicator of poor prognosis in patients with urothelial carcinoma treated with transurethral resection and radiochemotherapy. Virchows Arch 2016; 469:339-44. [PMID: 27392930 DOI: 10.1007/s00428-016-1986-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 05/27/2016] [Accepted: 07/03/2016] [Indexed: 10/21/2022]
Abstract
Purpose of this study was to evaluate prognostic impact of rare variants of urothelial bladder cancer (BC) after treatment with combined radiochemotherapy (RCT). To this end tumour tissue of 238 patients with urothelial carcinoma (UC) treated with transurethral resection of the bladder (TUR-B) and RCT with curative intent was collected. Histomorphological analysis included re-evaluation and semi-quantitative assessment of rare UC subtypes. Additionally, human epidermal growth factor receptor 2 (HER2) chromogenic in situ hybridisation (CISH) was performed in tumours with a micropapillary component exceeding 30 %. Long-term follow-up was available for 200 patients (range 3-282 months). Variant UC histology was found in 45 of 238 tumours, most frequently micropapillary UC (N = 17) including cases with a small fraction of tumour with micropapillary morphology. The mere presence of micropapillary morphology did not affect prognosis. In tumours with extensive (≥30 %) micropapillary morphology (N = 8) Kaplan-Meier analysis revealed significantly worse cancer specific survival (CSS) (P = 0.002) compared to conventional UC (mean survival times 97 months and 229 months, respectively). Univariate Cox regression analysis of cases with ≥30 % micropapillary morphology revealed a hazard ratio of 4.726 (95 % CI 1.629-13.714) for CSS (P = 0.004). CISH revealed HER2 gene amplification in 3/10 tumours with ≥30 % micropapillary component. In conclusion, for BC treated with TUR-B and RCT, the presence of micropapillary morphology in more than 30 % of the tumour is an adverse prognostic factor. Further studies are needed to evaluate a potential benefit of different, especially multimodal treatment strategies for micropapillary UC and also other subtypes of UC. Her2 represents a promising therapeutic target in a subset of micropapillary UC.
Collapse
|
35
|
HER2 Protein Overexpression and Gene Amplification in Plasmacytoid Urothelial Carcinoma of the Urinary Bladder. DISEASE MARKERS 2016; 2016:8463731. [PMID: 27034533 PMCID: PMC4806278 DOI: 10.1155/2016/8463731] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 02/18/2016] [Indexed: 01/06/2023]
Abstract
Aim. HER2 overexpression has been reported in a minority of urothelial carcinomas, but little is known about HER2 protein expression and gene alterations in plasmacytoid urothelial carcinoma, a rare and aggressive variant. The aim of this study was to clarify the HER2 status in plasmacytoid urothelial carcinomas. Methods. Six cases of plasmacytoid urothelial carcinoma were included, in which we evaluated HER2 protein expression by immunohistochemistry (IHC) and HER2 gene amplification by fluorescence in situ hybridization (FISH). Results. The patients' ages ranged from 57 to 83 years (mean age, 71 years). Five patients were male and one was female. The ratio of the plasmacytoid component ranged from 30% to 100% (mean, 77%). HER2 expression score was 3+ in 4 cases, 2+ in one case, and negative in one case. HER2 gene amplification was positive in 3 cases, of which 2 cases showed a 3+ HER2 IHC score but one case was negative for HER2 IHC. Another 2 cases showed equivocal HER2 FISH results, and one remaining case was negative for HER2 FISH. Conclusion. Our observation that plasmacytoid urothelial carcinomas frequently demonstrated HER2 protein overexpression provides supporting evidence that HER2 may be a potential therapeutic target for plasmacytoid urothelial carcinoma.
Collapse
|
36
|
Using Copy Number Alterations to Identify New Therapeutic Targets for Bladder Carcinoma. Int J Mol Sci 2016; 17:271. [PMID: 26927059 PMCID: PMC4813135 DOI: 10.3390/ijms17030271] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 02/17/2016] [Accepted: 02/17/2016] [Indexed: 01/25/2023] Open
Abstract
Bladder cancer represents the ninth most widespread malignancy throughout the world. It is characterized by the presence of two different clinical and prognostic subtypes: non-muscle-invasive bladder cancers (NMIBCs) and muscle-invasive bladder cancers (MIBCs). MIBCs have a poor outcome with a common progression to metastasis. Despite improvements in knowledge, treatment has not advanced significantly in recent years, with the absence of new therapeutic targets. Because of the limitations of current therapeutic options, the greater challenge will be to identify biomarkers for clinical application. For this reason, we compared our array comparative genomic hybridization (array-CGH) results with those reported in literature for invasive bladder tumors and, in particular, we focused on the evaluation of copy number alterations (CNAs) present in biopsies and retained in the corresponding cancer stem cell (CSC) subpopulations that should be the main target of therapy. According to our data, CCNE1, MYC, MDM2 and PPARG genes could be interesting therapeutic targets for bladder CSC subpopulations. Surprisingly, HER2 copy number gains are not retained in bladder CSCs, making the gene-targeted therapy less interesting than the others. These results provide precious advice for further study on bladder therapy; however, the clinical importance of these results should be explored.
Collapse
|
37
|
Ide H, Miyamoto H. Steroid Hormone Receptor Signals as Prognosticators for Urothelial Tumor. DISEASE MARKERS 2015; 2015:840640. [PMID: 26770009 PMCID: PMC4685115 DOI: 10.1155/2015/840640] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 11/18/2015] [Indexed: 12/18/2022]
Abstract
There is a substantial amount of preclinical or clinical evidence suggesting that steroid hormone receptor-mediated signals play a critical role in urothelial tumorigenesis and tumor progression. These receptors include androgen receptor, estrogen receptors, glucocorticoid receptor, progesterone receptor, vitamin D receptor, retinoid receptors, peroxisome proliferator-activated receptors, and others including orphan receptors. In particular, studies using urothelial cancer tissue specimens have demonstrated that elevated or reduced expression of these receptors as well as alterations of their upstream or downstream pathways correlates with patient outcomes. This review summarizes and discusses available data suggesting that steroid hormone receptors and related signals serve as biomarkers for urothelial carcinoma and are able to predict tumor recurrence or progression.
Collapse
Affiliation(s)
- Hiroki Ide
- Departments of Pathology and Urology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Hiroshi Miyamoto
- Departments of Pathology and Urology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
38
|
Sun Z, Shi Y, Shen Y, Cao L, Zhang W, Guan X. Analysis of different HER-2 mutations in breast cancer progression and drug resistance. J Cell Mol Med 2015; 19:2691-701. [PMID: 26305917 PMCID: PMC4687700 DOI: 10.1111/jcmm.12662] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 07/03/2015] [Indexed: 12/17/2022] Open
Abstract
Studies over the last two decades have identified that amplified human epidermal growth factor receptor (HER‐2; c‐erbB‐2, neu) and its overexpression have been frequently implicated in the carcinogenesis and prognosis in a variety of solid tumours, especially breast cancer. Lots of painstaking efforts were invested on the HER‐2 targeted agents, and significantly improved outcome and prolonged the survival of patients. However, some patients classified as ‘HER‐2‐positive’ would be still resistant to the anti‐HER‐2 therapy. Various mechanisms of drug resistance have been illustrated and the alteration of HER‐2 was considered as a crucial mechanism. However, systematic researches in regard to the HER‐2 mutations and variants are still inadequate. Notably, the alterations of HER‐2 play an important role in drug resistance, but also have a potential association with the cancer risk. In this review, we summarize the possible mutations and focus on HER‐2 variants’ role in breast cancer tumourigenesis. Additionally, the alteration of HER‐2, as a potential mechanism of resistance to trastuzumab, is discussed here. We hope that HER‐2 related activating mutations could potentially offer more therapeutic opportunities to a broader range of patients than previously classified as HER‐2 overexpressed.
Collapse
Affiliation(s)
- Zijia Sun
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yaqin Shi
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yan Shen
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lulu Cao
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wenwen Zhang
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiaoxiang Guan
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.,Department of Medical Oncology, Jinling Hospital, School of Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|