Review
Published: 2023-01-25
download
PDF

News in the classification of WHO 2022 bladder tumors

Histopathology and Molecular Diagnostics, University Hospital Careggi, Florence, Florence, Italy
Department of Pathology, Medical University of Vienna, General Hospital of Vienna, Wien, AT, Austria
University of Cordoba, Cordoba, Spain
Molecular Medicine and Cell Therapy Foundation, c/o Polytechnic University of the Marche Region, Ancona, Italy
Section of Pathological Anatomy, School of Medicine, University of Udine, Udine, Italy
Department of Surgical Pathology, Aichi Medical University, Aichi, Japan
Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
bladder urothelial tumors grading molecular subgroups WHO 2022

Abstract

The fifth-edition of World Health Organization (WHO) Classification of Tumors series for urinary and male genital tract tumors has been published, six years later the fourth-edition. In these years, new treatment approaches have been implemented and new molecular data on urological cancers are known. Morphology remains the groundwork for taxonomy of the urinary tract tumors. However, a molecular approach to classification of urothelial carcinomas and the management of selected neoplasms with new therapeutic modalities such as immunotherapy are emerging. More data are needed for the application of these advances in routine pathology practice and patient management. The 2022 World Health Organization (WHO) Classification of Tumors of the Urinary System and Male Genital Organs represents an update in classification on urinary tract tumors. It also offers new insights with regards to the grading of heterogeneous non-invasive urothelial neoplasms, the definition of inverted neoplasms, the grading of invasive urothelial carcinomas, the diversity of morphological appearance of urothelial carcinomas, the definition of precursor lesions and the lineage of differentiation of the tumors.

The grading

The grading of urotelial neoplasms is based on the level of architectural and cytological disorders. Low-grade carcinoma is characterized by lack of marked variation on nuclear size, mild loss of nuclear polarity and mild variation in nuclear size. High-grade carcinoma is characterized by marked architectural and cytologic disorders, loss of polarity, presence of irregular and pleomorphic nuclei, high number of mitoses and anaplasia.

The two-tier distinction between low-grade and high-grade carcinomas was first proposed in the 1998 WHO/ISUP consensus 1 and this has been promulgated by WHO in the third 2 and the fourth 3 edition and maintained in the fifth 4.

Low-grade and high-grade urothelial carcinomas are characterized by differences in prevalence of genetic alterations. The diagnosis of the two tumor groups (low- and high-grade) allows for correlation with cytological diagnosis using the terminology of the Paris System (TPS) 5. The TPS, focuses on identification of high-grade tumors, is clinical relevant and has been widely adopted. In addition, the use of low- and high-grade categories has better reproducibility and reduces the interobserver variability and is aligned with current patient management algorithms 6.

According to the 2022 classification for urinary tract neoplasia, every invasive urothelial carcinoma must be graded 7. In a substantial percentage of cases, invasive urothelial carcinomas are characterized by high-grade cytological findings. Uncommonly, invasive urothelial carcinomas lacking marked nuclear atypia and architectural disorder – both in exophytyc and invasive components – can be encountered. It is important to distinguishing low-grade papillary tumor with lamina propria invasion among the pT1 carcinomas; in fact, literature data show that invasive pT1 low-grade tumors reveal more favorable outcome than pT1 high-grade carcinomas (Fig. 1a, 1b) 8-10. Hence, clinical management schemes of pT1 urothelial carcinoma patients recognize different risk groups for low vs high-grade invasive carcinomas 11-14.

It is of note that subtypes of urothelial carcinoma and those with divergent differentiation are all considered high-grade tumors, even subtypes with a bland morphology, such as nested-type or microcystic type urothelial carcinomas 15-18.

From a pathological point of view, a critical issue is the heterogeneity of grade which occurs in one third of non-invasive papillary urothelial carcinomas, as grading is decisive for selecting the therapeutic approach. The lack of a clear cut-off for the distinguishing low- from high-grade carcinomas has led to variability in application of the criteria and sometimes poor interobserver reproducibility. Different proposals for better stratification of papillary urothelial carcinomam have been forwerded over the last years 19-21.

The Editorial Board of this fifth edition believes that there is a need to for better interobserver reproducibility for heterogeneous tumors and suggest to adopt common diagnostic criteria as a recent study has described 22. Therefore, the WHO 2022 blue book propose reporting papillary tumors as high-grade as long as the high-grade component represents 5% or more of the tumor. Moreover, tumors with less than 5% high-grade component should be reported as low-grade with less than 5% high-grade component. This clear approach for reporting, if widely adopted, will help to achieve more interobserver reproducibility and allow for better future correlation with clinical outcomes. In the future, machine learning and artificial intelligence might help pathologists in this regard. Multidisciplinary international collaboration for this issue is suggested.

Non-invasive urothelial lesions

Regarding non invasive papillary neoplasms, the WHO 2022 maintains the papillary urothelial neoplasia of low malignant potential (PUNLMP) which has lower rates of recurrence and progression than pTa LG carcinoma 23,24 as a clinically relevant category, as well as the low-grade, and the high-grade papillary carcinoma categories.

It is important to recognize the inverted pattern of growth and to differentiate it from the invasive lesions. The reporting of presence of inverted pattern of growth also allows for better correlation with cystoscopic findings. Clarifications criteria for the differential diagnosis for tumors with inverted histology have been itemized. This includes the non-invasive papillary urothelial carcinoma, papillary urothelial neoplasm of low malignant potential and urothelial papilloma groups. While dedicated sections are reserved for uncommon benign lesions such as urothelial papilloma and inverted urothelial papilloma, other inverted lesions (eg. Inverted PUNLMP and inverted low-grade and high-grade non-invasive carcinomas) are included in the sections dedicated to the more common exophytic counterparts 25-28.

Subtype of urothelial carcinoma

The terminology of histologic “subtypes” has replaced “variants” throughout the fifth series. Invasive urothelial carcinoma presents with a diversity of morphologic appearances (Figs. 2-9). The different subtypes of urothelial carcinoma can occur in pure forms or more commonly admixed with conventional urothelial carcinoma. There are new subtypes of urothelial carcinoma, such as the tubular urothelial carcinoma, which in pure fashion is caracterized by bland cells, arranged in tubular structures, and the large nested urothelial carcinoma 29-30 that has been distinguished from the nested type.

In addition there are refinements of the name clear cell urothelial carcinoma with the addition of the term glycogen rich 31-32 to clearly discriminate it from clear cell adenocarcinoma with Mullerian differentiation; and the term signet ring/diffuse has been dropped from the plasmacytoid subtype.

The knowledge of diversity in appearance of urothelial carcinoma with its specific subtypes is important for the recognition of the deceptively benign appearance of some subtypes such as microcystic, tubular, nested and large nested types when in pure form and for the differential diagnosis with non-urothelial tumor with morphological similarities.

Urothelial carcinoma can present areas of divergent differentiation that include glandular, squamous 33, trophoblastic 34, Mullerian 35 and even neuroendocrine lineages. The fifth edition underlines the need to report the presence and percentage(s) of the different subtypes and divergent differentiations in urothelial carcinomas which can be useful for patient management 36-38.

Precursor lesions

UROTHELIAL DYSPLASIA

The term dysplasia is controversial, not the least as a result of being based on the absence of criteria for a definite diagnosis of carcinoma in situ (CIS). Dysplasia in the urinary tract does not equate with CIS. The diagnosis of dysplasia consists of a preneoplastic lesion that “cytologically fall short of the diagnosis of carcinoma in situ39. The diagnostic term dysplasia is maintained in the fifth edition. However, instead of having its own entire chapter, like in the fourth-edition, it turns into a short description in the Urothelial Carcinoma in Situ chapter.

UROTHELIAL PAPILLARY PROLIFERATION OF UNDETERMINED MALIGNANT POTENTIAL

In the 2016 edition of the classification for urinary tract neoplasia, undulating urothelial proliferations that were considered like precursors of non-invasive low-grade papillary urothelial carcinoma were termed ”urothelial proliferation with undetermined malignant potential”. For such lesions, the term atypical urothelial proliferation (AUP) with tented and flat patterns has also been proposed 40; hovewer, in the 2022 edition, these lesions are no longer recognized as a stand-alone entity and are considered either early noninvasive low-grade papillary carcinomas or extensions of the edge of such tumors.

Organization of the fifth edition

Each tumor type is described listing benign and malignant entities. Urinary tract neoplasms are now listed in chapters that are mainly organized by tumor lineages of differentiation: urothelial, squamous and glandular tumors. Therefore, there are no longer dedicated organ-based chapters for urothelial tumors of prostate, urethra and upper urinary tract. Exceptions are made for histological findings useful for the diagnosis of rare tumors, such as urachal, diverticular and urethral accessory glands tumors that are described in specific sections.

Neuroendocrine neoplasms are described in a dedicated section. The classification which had been developed at the 2017 International Agency for Research on Cancer (IARC) neuroendocrine neoplasms consensus conference 41 is adopted for genitourinary tract organs as well as for all fifth-edition of World Health Organization (WHO) Classification of tumor series.

Mesenchymal, hematolymphoid, melanocitic and metastatic tumor are also reported in specific chapters. Genetic syndromes relevant for urinary tract tumors are described in the chapter on Genetic tumor syndromes of the urinary and male genital tract.

Unsolved matters

There remains some unsolved matters in bladder and urinary tract pathology in fifth-edition, such as the substaging for pT1 tumors and the molecular classification for urothelial carcinoma.

SUBSTAGING FOR PT1 TUMORS

Bladder cancer is staged using the TNM system 42-45. The depth of tumor invasion into bladder wall and adjacent structures defines the pT categories. Tumor stage represents one of the most important factors for prognosis and management in bladder cancer patients. An unresolved matter is the substaging for pT1 HG urothelial carcinoma although there is agreement on the prognostic value of the depth of lamina propria invasion 46-47. The fragmented nature of transurethral resection specimens and lack of orientation cause difficulties in the assessment of depth of lamina propria invasion. Histoanatomical and micrometric based systems have been proposed for substaging 48-56. Using a histoanatomical approach, the pT1 substage is evaluated according to tumor invasion within or beyond the muscularis mucosae or the large vessels of the lamina propria. The micrometric approach is based on the actual measurement of the invasive component. For the latter approach, different cut-offs have been proposed for substaging of invasion in the lamina propria. Despite the variability in the cut-off of micrometric approach and/or the type of approach to be used, the fifth edition strongly suggests to quantify tumor invasion in lamina propria with any of the proposed systems.

MOLECULAR FINDINGS

Recent studies on muscle-invasive bladder carcinoma have revealed six molecular subgroups with different prognosis 57-61. Luminal-papillary subgroups are identified in 24% of muscle-invasive tumors, luminal non-specified in 8%; luminal-unstable in 15%, stromal-rich in 15%, basal-squamous in 35% and neuroendocrine-like in 3% of muscle-invasive bladder carcinoma.

The various gene expression based subgroups are enriched for some histologic features, immune microenvironments and gene signatures. For example, FGFR3 mutations are more frequent in luminal-papillary subtype, while TP53 mutations are more likely in neuroendocrine-like, basal-squamous and luminal-unstable.

From a therapeutic point of view, alterations in ERCC2 and other DNA damage repair genes are have been associated with better response to cisplatin-based chemotherapy 62-63. Mutation, amplification, and fusion of FGFR3 can be associated with anti-FGFR agents 64-65.

Athough at the current time it is not possible to go towards a molecular classification of urinary tract tumors, the 2022 blue book highlights the great potential impact of the novel molecular advances for diagnosis and management of urinary tract tumors 66-68.

CONFLICTS OF INTEREST

The authors declare no conflict of interest.

MRR, EMC, TT and GJN served as expert members in 2022 WHO Classification of Tumours Editorial Board. Urinary and male genital tumours.

FUNDING

The authors declare no funding,

AUTHORS’ CONTRIBUTIONS

Writing-original draft preparation, MRR; writing-review and editing, MRR, EMC, ALB, RM, AC, TT, GJN. All authors have read and agreed to the published version of the manuscript.

Figures and tables

Figure 1a.Papillary urothelial carcinoma, low grade, with a focus of low-grade urothelial carcinoma invading lamina propria.

Figure 1b.Papillary urothelial carcinoma, high grade, invading lamina propria.

Figure 2a.Urothelial carcinoma with trophoblastic differentiation. Tumor with syncytiotrophoblasytic cells.

Figure 2b.Urothelial carcinoma with trophoblastic differentiation. Beta-hCG stain positive in syncytiotrophoblasytic cells.

Figure 3.Microcystic urothelial carcinoma. Presence of cystic structures infiltrating a non-reactive stroma.

Figure 4.Micropapillary urothelial carcinoma. Tumor with micropapillary architecture.

Figure 5a.Lymphoepithelioma-like urothelial carcinoma.

Figure 5b.Lymphoepithelioma-like urothelial carcinoma. Sheet of CK7 positive cells with a syncytial feature.

Figure 6.Lipid-rich urothelial carcinoma. Tumor cells with a lipoblast-like appearance.

Figure 7.Plasmacytoid urothelial carcinoma. Dyscoesive and plasmacytoid-appearance tumor cells invading the perivesical fat.

Figure 8.Clear cell (glycogen-rich) urothelial carcinoma. Sheet of cells with clear cytoplasm invading the muscle.

Figure 9.Sarcomatoid urothelial carcinoma. Spindle tumor cells with high grade atypia.

References

  1. Epstein JI, Amin MB, Reuter VR. The World Health Organization/International Society of Urological Pathology consensus classification of urothelial (transitional cell) neoplasms of the urinary bladder. Bladder Consensus Conference Committee. Am J Surg Pathol. 1998; 22:1435-1448. DOI | PubMed
  2. IARC Press: Lyon (France); 2004. Publisher Full Text
  3. IARC Press: Lyon (France); 2016. Publisher Full Text
  4. WHO Classification of Tumours Editorial Board. IARC Press: Lyon (France); 2022.
  5. The Paris System (TPS) for reporting cytology. The Paris System for reporting urinary cytology. Springer International Publishing: Cham (Switzerland); 2016.
  6. Sylvester RJ, Rodríguez O, Hernández V. European Association of Urology (EAU) Prognostic Factor Risk Groups for Non-muscle-invasive Bladder Cancer (NMIBC) Incorporating the WHO 2004/2016 and WHO 1973 Classification Systems for Grade: An Update from the EAU NMIBC Guidelines Panel. Eur Urol. 2021; 79:480-488. DOI | PubMed
  7. Amin MB, Smith SC, Reuter VE. Update for the practicing pathologist: the International Consultation On Urologic Disease-European association of urology consultation on bladder cancer. Mod Pathol. 2015; 28:612-630. DOI | PubMed |
  8. Pan CC, Chang YH, Chen KK. Constructing prognostic model incorporating the 2004 WHO/ISUP classification for patients with non-muscle-invasive urothelial tumours of the urinary bladder. J Clin Pathol. 2010; 63:910-915. DOI | PubMed
  9. Toll AD, Epstein JI. Invasive low-grade papillary urothelial carcinoma: a clinicopathologic analysis of 41 cases. Am J Surg Pathol. 2012; 36:1081-1086. DOI | PubMed
  10. Holmäng S, Andius P, Hedelin H. Stage progression in Ta papillary urothelial tumors: relationship to grade, immunohistochemical expression of tumor markers, mitotic frequency and DNA ploidy. J Urol. 2001; 165:1124-1128. PubMed
  11. Babjuk M, Burger M, Compérat EM. European Association of Urology Guidelines on Non-muscle-invasive Bladder Cancer (TaT1 and Carcinoma In Situ) - 2019 Update. Eur Urol. 2019; 76:639-657. DOI | PubMed
  12. Soukup V, Čapoun O, Cohen D. Risk Stratification Tools and Prognostic Models in Non-muscle-invasive Bladder Cancer: A Critical Assessment from the European Association of Urology Non-muscle-invasive Bladder Cancer Guidelines Panel. Eur Urol Focus. 2020; 6:479-489. DOI | PubMed
  13. Chang SS, Boorjian SA, Chou R. Diagnosis and Treatment of Non-Muscle Invasive Bladder Cancer: AUA/SUO Guideline. J Urol. 2016; 196:1021-1029. DOI | PubMed
  14. Babjuk M, Burger M, Compérat E. EAU Guidelines Office: Arnhem (Netherlands); 2021. Publisher Full Text
  15. Beltran AL, Cheng L, Montironi R. Clinicopathological characteristics and outcome of nested carcinoma of the urinary bladder. Virchows Arch. 2014; 465:199-205. DOI | PubMed
  16. Drew PA, Furman J, Civantos F. The nested variant of transitional cell carcinoma: an aggressive neoplasm with innocuous histology. Mod Pathol. 1996; 9:989-994. PubMed
  17. Lopez Beltran A, Montironi R, Cheng L. Microcystic urothelial carcinoma: morphology, immunohistochemistry and clinical behaviour. Histopathology. 2014; 64:872-879. DOI | PubMed
  18. Veskimäe E, Espinos EL, Bruins HM. What Is the Prognostic and Clinical Importance of Urothelial and Nonurothelial Histological Variants of Bladder Cancer in Predicting Oncological Outcomes in Patients with Muscle-invasive and Metastatic Bladder Cancer? A European Association of Urology Muscle Invasive and Metastatic Bladder Cancer Guidelines Panel Systematic Review. Eur Urol Oncol. 2019; 2:625-642. DOI | PubMed
  19. Cheng L, Neumann RM, Nehra A. Cancer heterogeneity and its biologic implications in the grading of urothelial carcinoma. Cancer. 2000; 88:1663-1670. DOI | PubMed
  20. Gofrit ON, Pizov G, Shapiro A. Mixed high and low grade bladder tumors--are they clinically high or low grade?. J Urol. 2014; 191:1693-1696. DOI | PubMed
  21. Schubert T, Danzig MR, Kotamarti S. Mixed low- and high-grade non-muscle-invasive bladder cancer: a histological subtype with favorable outcome. World J Urol. 2015; 33:847-852. DOI | PubMed
  22. Reis LO, Taheri D, Chaux A. Significance of a minor high-grade component in a low-grade noninvasive papillary urothelial carcinoma of bladder. Hum Pathol. 2016; 47:20-25. DOI | PubMed
  23. Holmäng S, Hedelin H, Anderström C. Recurrence and progression in low grade papillary urothelial tumors. J Urol. 1999; 162(3 Pt 1):702-707. DOI | PubMed
  24. Holmäng S, Andius P, Hedelin H. Stage progression in Ta papillary urothelial tumors: relationship to grade, immunohistochemical expression of tumor markers, mitotic frequency and DNA ploidy. J Urol. 2001; 165:1124-1128. PubMed
  25. Amin MB, Gómez JA, Young RH. Urothelial transitional cell carcinoma with endophytic growth patterns: a discussion of patterns of invasion and problems associated with assessment of invasion in 18 cases. Am J Surg Pathol. 1997; 21:1057-1068. DOI | PubMed
  26. Hodges KB, Lopez-Beltran A, Maclennan GT. Urothelial lesions with inverted growth patterns: histogenesis, molecular genetic findings, differential diagnosis and clinical management. BJU Int. 2011; 107:532-537. DOI | PubMed
  27. Amin MB, Smith SC, Reuter VE. Update for the practicing pathologist: The International Consultation On Urologic Disease-European association of urology consultation on bladder cancer. Mod Pathol. 2015; 28:612-630. DOI | PubMed |
  28. Guo A, Liu A, Teng X. The pathology of urinary bladder lesions with an inverted growth pattern. Chin J Cancer Res. 2016; 28:107-121. DOI | PubMed |
  29. Compérat E, McKenney JK, Hartmann A. Large nested variant of urothelial carcinoma: a clinicopathological study of 36 cases. Histopathology. 2017; 71:703-710. DOI | PubMed
  30. Cox R, Epstein JI. Large nested variant of urothelial carcinoma: 23 cases mimicking von Brunn nests and inverted growth pattern of noninvasive papillary urothelial carcinoma. Am J Surg Pathol. 2011; 35:1337-1342. DOI | PubMed
  31. Zhou Z, Kinslow CJ, Wang P. Clear Cell Adenocarcinoma of the Urinary Bladder Is a Glycogen-Rich Tumor with Poorer Prognosis. J Clin Med. 2020; 9:138. DOI | PubMed |
  32. Chan EO, Chan VW, Poon JY. Clear cell carcinoma of the urinary bladder: a systematic review. Int Urol Nephrol. 2021; 53:815-824. DOI | PubMed
  33. Kim SP, Frank I, Cheville JC. The impact of squamous and glandular differentiation on survival after radical cystectomy for urothelial carcinoma. J Urol. 2012; 188:405-409. DOI | PubMed
  34. Przybycin CG, McKenney JK, Nguyen JK. Urothelial Carcinomas With Trophoblastic Differentiation, Including Choriocarcinoma: Clinicopathologic Series of 16 Cases. Am J Surg Pathol. 2020; 44:1322-1330. DOI | PubMed
  35. Herawi M, Drew PA, Pan CC. Clear cell adenocarcinoma of the bladder and urethra: cases diffusely mimicking nephrogenic adenoma. Hum Pathol. 2010; 41:594-601. DOI | PubMed
  36. Takahara T, Murase Y, Tsuzuki T. Urothelial carcinoma: variant histology, molecular subtyping, and immunophenotyping significant for treatment outcomes. Pathology. 2021; 53:56-66. DOI | PubMed
  37. Mori K, Abufaraj M, Mostafaei H. A Systematic Review and Meta-Analysis of Variant Histology in Urothelial Carcinoma of the Bladder Treated with Radical Cystectomy. J Urol. 2020; 204:1129-1140. DOI | PubMed
  38. Moschini M, D’Andrea D, Korn S. Characteristics and clinical significance of histological variants of bladder cancer. Nat Rev Urol. 2017; 14:651-668. DOI | PubMed
  39. McKenney JK. Precursor lesions of the urinary bladder. Histopathology. 2019; 74:68-76. DOI | PubMed
  40. Amin MB, Comperat E, Epstein JI. The Genitourinary Pathology Society Update on Classification and Grading of Flat and Papillary Urothelial Neoplasia With New Reporting Recommendations and Approach to Lesions With Mixed and Early Patterns of Neoplasia. Adv Anat Pathol. 2021; 28:179-195. DOI | PubMed
  41. Rindi G, Klimstra DS, Abedi-Ardekani B. A common classification framework for neuroendocrine neoplasms: an International Agency for Research on Cancer (IARC) and World Health Organization (WHO) expert consensus proposal. Mod Pathol. 2018; 31:1770-1786. DOI | PubMed |
  42. Paner GP, Stadler WM, Hansel DE. Updates in the Eighth Edition of the Tumor-Node-Metastasis Staging Classification for Urologic Cancers. Eur Urol. 2018; 73:560-569. DOI | PubMed
  43. Paner GP, Montironi R, Amin MB. Challenges in Pathologic Staging of Bladder Cancer: Proposals for Fresh Approaches of Assessing Pathologic Stage in Light of Recent Studies and Observations Pertaining to Bladder Histoanatomic Variances. Adv Anat Pathol. 2017; 24:113-127. DOI | PubMed
  44. Magers MJ, Lopez-Beltran A, Montironi R. Staging of bladder cancer. Histopathology. 2019; 74:112-134. DOI | PubMed
  45. Gallan AJ, Choy B, Paner GP. Contemporary Grading and Staging of Urothelial Neoplasms of the Urinary Bladder: New Concepts and Approaches to Challenging Scenarios. Surg Pathol Clin. 2018; 11:775-795. DOI | PubMed
  46. Kardoust Parizi M, Enikeev D, Glybochko PV. Prognostic value of T1 substaging on oncological outcomes in patients with non-muscle-invasive bladder urothelial carcinoma: a systematic literature review and meta-analysis. World J Urol. 2020; 38:1437-1449. DOI | PubMed |
  47. Lawless M, Gulati R, Tretiakova M. Stalk versus base invasion in pT1 papillary cancers of the bladder: improved substaging system predicting the risk of progression. Histopathology. 2017; 71:406-414. DOI | PubMed |
  48. Raspollini MR, Montironi R, Mazzucchelli R. pT1 high-grade bladder cancer: histologic criteria, pitfalls in the assessment of invasion, and substaging. Virchows Arch. 2020; 477:3-16. DOI | PubMed
  49. Colombo R, Hurle R, Moschini M. Feasibility and Clinical Roles of Different Substaging Systems at First and Second Transurethral Resection in Patients with T1 High-Grade Bladder Cancer. Eur Urol Focus. 2018; 4:87-93. DOI | PubMed
  50. Fransen van de Putte EE, Otto W, Hartmann A. Metric substage according to micro and extensive lamina propria invasion improves prognostics in T1 bladder cancer. Urol Oncol. 2018; 36:361.e7-361.e13. DOI | PubMed
  51. Chang WC, Chang YH, Pan CC. Prognostic significance in substaging ofT1 urinary bladder urothelial carcinoma on transurethral resection. Am J Surg Pathol. 2012; 36:454-461. DOI | PubMed
  52. van Rhijn BW, van der Kwast TH, Alkhateeb SS. A new and highly prognostic system to discern T1 bladder cancer substage. Eur Urol. 2012; 61:378-384. DOI | PubMed
  53. Hu Z, Mudaliar K, Quek ML. Measuring the dimension of invasive component in pT1 urothelial carcinoma in transurethral resection specimens can predict time to recurrence. Ann Diagn Pathol. 2014; 18:49-52. DOI | PubMed
  54. Bertz S, Denzinger S, Otto W. Substaging by estimating the size of invasive tumour can improve risk stratification in pT1 urothelial bladder cancer-evaluation of a large hospital-based single-centre series. Histopathology. 2011; 59:722-732. DOI | PubMed
  55. Leivo MZ, Sahoo D, Hamilton Z. Analysis of T1 Bladder Cancer on Biopsy and Transurethral Resection Specimens: Comparison and Ranking of T1 Quantification Approaches to Predict Progression to Muscularis Propria Invasion. Am J Surg Pathol. 2018; 42:e1-e10. DOI | PubMed
  56. Paner GP, Montironi R, Amin MB. Challenges in Pathologic Staging of Bladder Cancer: Proposals for Fresh Approaches of Assessing Pathologic Stage in Light of Recent Studies and Observations Pertaining to Bladder Histoanatomic Variances. Adv Anat Pathol. 2017; 24:113-127. DOI | PubMed
  57. Kamoun A, de Reyniès A, Allory Y. A Consensus Molecular Classification of Muscle-invasive Bladder Cancer. Eur Urol. 2020; 77:420-433. DOI | PubMed |
  58. Sjödahl G, Lövgren K, Lauss M. Toward a molecular pathologic classification of urothelial carcinoma. Am J Pathol. 2013; 183:681-691. DOI | PubMed
  59. Choi W, Porten S, Kim S. Identification of distinct basal and luminal subtypes of muscle-invasive bladder cancer with different sensitivities to frontline chemotherapy. Cancer Cell. 2014; 25:152-165. DOI | PubMed |
  60. Robertson AG, Kim J, Al-Ahmadie H. Comprehensive Molecular Characterization of Muscle-Invasive. Bladder Cancer Cell. 2017; 171:540-556.e25. DOI | PubMed |
  61. Mo Q, Nikolos F, Chen F. Prognostic Power of a Tumor Differentiation Gene Signature for Bladder Urothelial Carcinomas. J Natl Cancer Inst. 2018; 110:448-459. DOI | PubMed |
  62. Van Allen EM, Mouw KW, Kim P. Somatic ERCC2 mutations correlate with cisplatin sensitivity in muscle-invasive urothelial carcinoma. Cancer Discov. 2014; 4:1140-1153. DOI | PubMed |
  63. Iyer G, Balar AV, Milowsky MI. Multicenter Prospective Phase II Trial of Neoadjuvant Dose-Dense Gemcitabine Plus Cisplatin in Patients With Muscle-Invasive Bladder Cancer. J Clin Oncol. 2018; 36:1949-1956. DOI | PubMed |
  64. Pal SK, Rosenberg JE, Hoffman-Censits JH. Efficacy of BGJ398, a Fibroblast Growth Factor Receptor 1-3 Inhibitor, in Patients with Previously Treated Advanced Urothelial Carcinoma with FGFR3 Alterations. Cancer Discov. 2018; 8:812-821. DOI | PubMed |
  65. Loriot Y, Necchi A, Park SH. Erdafitinib in Locally Advanced or Metastatic Urothelial Carcinoma. N Engl J Med. 2019; 381:338-348. DOI | PubMed
  66. Hodgson A, Slodkowska E, Jungbluth A. PD-L1 Immunohistochemistry Assay Concordance in Urothelial Carcinoma of the Bladder and Hypopharyngeal Squamous Cell Carcinoma. Am J Surg Pathol. 2018; 42:1059-1066. DOI | PubMed |
  67. Tretiakova M, Fulton R, Kocherginsky M. Concordance study of PD-L1 expression in primary and metastatic bladder carcinomas: comparison of four commonly used antibodies and RNA expression. Mod Pathol. 2018; 31:623-632. DOI | PubMed
  68. Reis H, Serrette R, Posada J. PD-L1 Expression in Urothelial Carcinoma With Predominant or Pure Variant Histology: Concordance Among 3 Commonly Used and Commercially Available Antibodies. Am J Surg Pathol. 2019; 43:920-927. DOI | PubMed |

Affiliations

$authorString->getOrcid() =>

$authorString->getFullName() => Maria Rosaria Raspollini

$authorString->getUrl() => https://orcid.org/0000-0001-6630-4881

Maria Rosaria Raspollini

Histopathology and Molecular Diagnostics, University Hospital Careggi, Florence, Florence, Italy
non esiste orcidID ""https://orcid.org/0000-0001-6630-4881

$authorString->getOrcid() =>

$authorString->getFullName() => Eva M. Comperat

$authorString->getUrl() =>

Eva M. Comperat

Department of Pathology, Medical University of Vienna, General Hospital of Vienna, Wien, AT, Austria
non esiste orcidID ""

$authorString->getOrcid() =>

$authorString->getFullName() => Antonio Lopez-Beltran

$authorString->getUrl() =>

Antonio Lopez-Beltran

University of Cordoba, Cordoba, Spain
non esiste orcidID ""

$authorString->getOrcid() =>

$authorString->getFullName() => Rodolfo Montironi

$authorString->getUrl() =>

Rodolfo Montironi

Molecular Medicine and Cell Therapy Foundation, c/o Polytechnic University of the Marche Region, Ancona, Italy
non esiste orcidID ""

$authorString->getOrcid() =>

$authorString->getFullName() => Alessia Cimadamore

$authorString->getUrl() =>

Alessia Cimadamore

Section of Pathological Anatomy, School of Medicine, University of Udine, Udine, Italy
non esiste orcidID ""

$authorString->getOrcid() =>

$authorString->getFullName() => Toyonori Tsuzuki

$authorString->getUrl() =>

Toyonori Tsuzuki

Department of Surgical Pathology, Aichi Medical University, Aichi, Japan
non esiste orcidID ""

$authorString->getOrcid() =>

$authorString->getFullName() => George J. Netto

$authorString->getUrl() =>

George J. Netto

Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
non esiste orcidID ""

Copyright

© Società Italiana di Anatomia Patologica e Citopatologia Diagnostica, Divisione Italiana della International Academy of Pathology , 2023

How to Cite

[1]
Raspollini, M.R., Comperat, E.M., Lopez-Beltran, A., Montironi, R., Cimadamore, A., Tsuzuki, T. and Netto, G.J. 2023. News in the classification of WHO 2022 bladder tumors. Pathologica - Journal of the Italian Society of Anatomic Pathology and Diagnostic Cytopathology. 115, 1 (Jan. 2023), 32-40. DOI:https://doi.org/10.32074/1591-951X-838.
  • Abstract viewed - 2242 times
  • PDF downloaded - 2990 times