Better prognostic markers for nonmuscle invasive papillary urothelial carcinomas

Authors

Keywords:

bladder cancer, blærekreft, papillary urothelial carcinomas, prognostic biomarkers, non-muscle invasive bladder cancer, immunotherapy

Synopsis

Bladder cancer is a common type of cancer, especially among men in developed countries. Most cancers in the urinary bladder are papillary urothelial carcinomas. They are characterized by a high recurrence frequency (up to 70 %) after local resection. It is crucial for prognosis to discover these recurrent tumours at an early stage, especially before they become muscle-invasive. Reliable prognostic biomarkers for tumour recurrence and stage progression are lacking. This is why patients diagnosed with a non-muscle invasive bladder cancer follow extensive follow-up regimens with possible serious side effects and with high costs for the healthcare systems.

WHO grade and tumour stage are two central biomarkers currently having great impact on both treatment decisions and follow-up regimens. However, there are concerns regarding the reproducibility of WHO grading, and stage classification is challenging in small and fragmented tumour material. In Paper I, we examined the reproducibility and the prognostic value of all the individual microscopic features making up the WHO grading system. Among thirteen extracted features there was considerable variation in both reproducibility and prognostic value. The only feature being both reasonably reproducible and statistically significant prognostic was cell polarity. We concluded that further validation studies are needed on these features, and that future grading systems should be based on well-defined features with true prognostic value.

With the implementation of immunotherapy, there is increasing interest in tumour immune response and the tumour microenvironment. In a search for better prognostic biomarkers for tumour recurrence and stage progression, in Paper II, we investigated the prognostic value of tumour infiltrating immune cells (CD4, CD8, CD25 and CD138) and previously investigated cell proliferation markers (Ki-67, PPH3 and MAI). Low Ki 67 and tumour multifocality were associated with increased recurrence risk. Recurrence risk was not affected by the composition of immune cells. For stage progression, the only prognostic immune cell marker was CD25. High values for MAI was also strongly associated with stage progression. However, in a multivariate analysis, the most prognostic feature was a combination of MAI and CD25.

BCG-instillations in the bladder are indicated in intermediate and high-risk non-muscle invasive bladder cancer patients. This old-fashion immunotherapy has proved to reduce both recurrence- and progression-risk, although it is frequently followed by unpleasant side-effects. As many as 30-50% of high-risk patients receiving BCG instillations, fail by develop high-grade recurrences. They do not only suffer from unnecessary side-effects, but will also have a delay in further treatment. Together with colleagues at three different Dutch hospitals, in Paper III, we looked at the prognostic and predictive value of T1-substaging. A T1-tumour invades the lamina propria, and we wanted to separate those with micro- from those with extensive invasion. We found that BCG-failure was more common among patients with extensive invasion. Furthermore, T1-substaging was associated with both high-grade recurrence-free and progression-free survival.

Finally, in Paper IV, we wanted to investigate the prognostic value of two classical immunohistochemical markers, p53 and CK20, and compare them with previously investigated proliferation markers. p53 is a surrogate marker for mutations in the gene TP53, considered to be a main characteristic for muscle-invasive tumours. CK20 is a surrogate marker for luminal tumours in the molecular classification of bladder cancer, and is frequently used to distinguish reactive urothelial changes from urothelial carcinoma in situ. We found both positivity for p53 and CK20 to be significantly associated with stage progression, although not performing better than WHO grade and stage. The proliferation marker MAI, had the highest prognostic value in our study. Any combination of variables did not perform better in a multivariate analysis than MAI alone.

Author Biography

Vebjørn Kvikstad

Phd fellow / Pathologist
Faculty of Science and Technology
Department of Chemistry, Bioscience and Environmental Engineering
University of Stavanger
vebjorn.kvikstad@sus.no

References

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394-424.

https://doi.org/10.3322/caac.21492

Kirkali Z, Chan T, Manoharan M, Algaba F, Busch C, Cheng L, et al. Bladder cancer: epidemiology, staging and grading, and diagnosis. Urology. 2005;66(6 Suppl 1):4-34.

https://doi.org/10.1016/j.urology.2005.07.062

Norway Cro. Cancer in Norway 2019 - Cancer incidence, mortality, survival and prevalence in Norway. 2020.

Moch H, Humphrey PA, Ulbright TM, Reuter VE. World Health Organization Classification of tumours. 2016:77 - 135.

Sievert KD, Amend B, Nagele U, Schilling D, Bedke J, Horstmann M, et al. Economic aspects of bladder cancer: what are the benefits and costs? World J Urol. 2009;27(3):295-300.

https://doi.org/10.1007/s00345-009-0395-z

Richters A, Aben KKH, Kiemeney L. The global burden of urinary bladder cancer: an update. World J Urol. 2020;38(8):1895-904.

https://doi.org/10.1007/s00345-019-02984-4

Dobruch J, Daneshmand S, Fisch M, Lotan Y, Noon AP, Resnick MJ, et al. Gender and Bladder Cancer: A Collaborative Review of Etiology, Biology, and Outcomes. Eur Urol. 2016;69(2):300-10.

https://doi.org/10.1016/j.eururo.2015.08.037

Cumberbatch MG, Rota M, Catto JW, La Vecchia C. The Role of Tobacco Smoke in Bladder and Kidney Carcinogenesis: A Comparison of Exposures and Meta-analysis of Incidence and Mortality Risks. Eur Urol. 2016;70(3):458-66.

https://doi.org/10.1016/j.eururo.2015.06.042

Chen CH, Shun CT, Huang KH, Huang CY, Tsai YC, Yu HJ, et al. Stopping smoking might reduce tumour recurrence in nonmuscle- invasive bladder cancer. BJU Int. 2007;100(2):281-6; discussion 6.

https://doi.org/10.1111/j.1464-410X.2007.06873.x

Afshari M, Janbabaei G, Bahrami MA, Moosazadeh M. Opium and bladder cancer: A systematic review and meta-analysis of the odds ratios for opium use and the risk of bladder cancer. PLoS One. 2017;12(6):e0178527.

https://doi.org/10.1371/journal.pone.0178527

Thomas AA, Wallner LP, Quinn VP, Slezak J, Van Den Eeden SK, Chien GW, et al. Association between cannabis use and the risk of bladder cancer: results from the California Men's Health Study. Urology. 2015;85(2):388-92.

https://doi.org/10.1016/j.urology.2014.08.060

Cumberbatch MGK, Jubber I, Black PC, Esperto F, Figueroa JD, Kamat AM, et al. Epidemiology of Bladder Cancer: A Systematic Review and Contemporary Update of Risk Factors in 2018. Eur Urol. 2018;74(6):784-95.

https://doi.org/10.1016/j.eururo.2018.09.001

Figueroa JD, Koutros S, Colt JS, Kogevinas M, Garcia-Closas M, Real FX, et al. Modification of Occupational Exposures on Bladder Cancer Risk by Common Genetic Polymorphisms. J Natl Cancer Inst. 2015;107(11).

https://doi.org/10.1093/jnci/djv223

Lukas C, Selinski S, Prager HM, Blaszkewicz M, Hengstler JG, Golka K. Occupational bladder cancer: Polymorphisms of xenobiotic metabolizing enzymes, exposures, and prognosis. J Toxicol Environ Health A. 2017;80(7-8):439-52.

https://doi.org/10.1080/10937404.2017.1304731

Garcia-Closas M, Rothman N, Figueroa JD, Prokunina-Olsson L, Han SS, Baris D, et al. Common genetic polymorphisms modify the effect of smoking on absolute risk of bladder cancer. Cancer Res. 2013;73(7):2211-20.

https://doi.org/10.1158/0008-5472.CAN-12-2388

Botteri E, Ferrari P, Roswall N, Tjønneland A, Hjartåker A, Huerta JM, et al. Alcohol consumption and risk of urothelial cell bladder cancer in the European prospective investigation into cancer and nutrition cohort. Int J Cancer. 2017;141(10):1963-70.

https://doi.org/10.1002/ijc.30894

Baris D, Waddell R, Beane Freeman LE, Schwenn M, Colt JS, Ayotte JD, et al. Elevated Bladder Cancer in Northern New England: The Role of Drinking Water and Arsenic. J Natl Cancer Inst. 2016;108(9).

https://doi.org/10.1093/jnci/djw099

Mendez WM, Jr., Eftim S, Cohen J, Warren I, Cowden J, Lee JS, et al. Relationships between arsenic concentrations in drinking water and lung and bladder cancer incidence in U.S. counties. J Expo Sci Environ Epidemiol. 2017;27(3):235-43.

https://doi.org/10.1038/jes.2016.58

Wallis CJ, Mahar AL, Choo R, Herschorn S, Kodama RT, Shah PS, et al. Second malignancies after radiotherapy for prostate cancer: systematic review and meta-analysis. BMJ. 2016;352:i851.

https://doi.org/10.1136/bmj.i851

Abern MR, Dude AM, Tsivian M, Coogan CL. The characteristics of bladder cancer after radiotherapy for prostate cancer. Urol Oncol. 2013;31(8):1628-34.

https://doi.org/10.1016/j.urolonc.2012.04.006

Lim A, Rao P, Matin SF. Lynch syndrome and urologic malignancies: a contemporary review. Current opinion in urology. 2019;29(4):357-63.

https://doi.org/10.1097/MOU.0000000000000639

Gripp KW, Rauen KA. Costello Syndrome. In: Adam MP, Ardinger HH, Pagon RA, Wallace SE, Bean LJH, Mirzaa G, et al., editors. GeneReviews(®). Seattle (WA): University of Washington, Seattle Copyright © 1993-2021, University of Washington, Seattle. GeneReviews is a registered trademark of the University of Washington, Seattle. All rights reserved.; 1993.

Zaghloul MS, Zaghloul TM, Bishr MK, Baumann BC. Urinary schistosomiasis and the associated bladder cancer: update. J Egypt Natl Canc Inst. 2020;32(1):44.

https://doi.org/10.1186/s43046-020-00055-z

Pasin E, Josephson DY, Mitra AP, Cote RJ, Stein JP. Superficial bladder cancer: an update on etiology, molecular development, classification, and natural history. Rev Urol. 2008;10(1):31-43.

Mostofi FKS, L. H.; Torloni, H. Histologic typing of urinary bladder tumours. Geneva: World Health Organization; 1973.

Moch H, Humphrey P, Ulbright T, Reuter V. WHO Classification of tumours of the urinary system and male genital organs. 4 ed. Lyon: International agency for research on cancer; 2016. p. 99 - 108.

Epstein JI, Amin MB, Reuter VR, Mostofi FK. 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(12):1435-48.

https://doi.org/10.1097/00000478-199812000-00001

EAU Guidelines. Edn. presented at the EAU Annual Congress Milan 2021. 2021 [

Helsedirektoratet. Nasjonalt handlingsprogram med retningslinjer for diagnostikk, behandling og oppfølging av blærekreft 2018 [updated 14.04.2021.

Kim JK, Moon KC, Jeong CW, Kwak C, Kim HH, Ku JH. Papillary Urothelial Neoplasm of Low Malignant Potential (PUNLMP) After Initial TUR-BT: Comparative Analyses with Noninvasive Low- Grade Papillary Urothelial Carcinoma (LGPUC). J Cancer. 2017;8(15):2885-91.

https://doi.org/10.7150/jca.20003

Sylvester RJ, Rodríguez O, Hernández V, Turturica D, Bauerová L, Bruins HM, et al. 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(4):480-8.

https://doi.org/10.1016/j.eururo.2020.12.033

Hentschel AE, van Rhijn BWG, Bründl J, Compérat EM, Plass K, Rodríguez O, et al. Papillary urothelial neoplasm of low malignant potential (PUN-LMP): Still a meaningful histo-pathological grade category for Ta, noninvasive bladder tumors in 2019? Urol Oncol. 2020;38(5):440-8.

https://doi.org/10.1016/j.urolonc.2019.10.002

Cheng L, Neumann RM, Nehra A, Spotts BE, Weaver AL, Bostwick DG. Cancer heterogeneity and its biologic implications in the grading of urothelial carcinoma. Cancer. 2000;88(7):1663-70.

https://doi.org/10.1002/(SICI)1097-0142(20000401)88:7<1663::AID-CNCR21>3.0.CO;2-8

Lopez-Beltran A, Montironi R. Non-invasive urothelial neoplasms: according to the most recent WHO classification. Eur Urol. 2004;46(2):170-6.

https://doi.org/10.1016/j.eururo.2004.03.017

Soukup V, Capoun O, Cohen D, Hernandez V, Babjuk M, Burger M, et al. Prognostic Performance and Reproducibility of the 1973 and 2004/2016 World Health Organization Grading Classification Systems in Non-muscle-invasive Bladder Cancer: A European Association of Urology Non-muscle Invasive Bladder Cancer Guidelines Panel Systematic Review. Eur Urol. 2017.

https://doi.org/10.1016/j.eururo.2017.04.015

van Rhijn BWG, Hentschel AE, Bründl J, Compérat EM, Hernández V, Čapoun O, et al. Prognostic Value of the WHO1973 and WHO2004/2016 Classification Systems for Grade in Primary Ta/T1 Non-muscle-invasive Bladder Cancer: A Multicenter European Association of Urology Non-muscle-invasive Bladder Cancer Guidelines Panel Study. European urology oncology. 2021;4(2):182-91.

https://doi.org/10.1016/j.euo.2020.12.002

van Rhijn BW, van Leenders GJ, Ooms BC, Kirkels WJ, Zlotta AR, Boevé ER, et al. The pathologist's mean grade is constant and individualizes the prognostic value of bladder cancer grading. Eur Urol. 2010;57(6):1052-7.

https://doi.org/10.1016/j.eururo.2009.09.022

Yorukoglu K, Tuna B, Dikicioglu E, Duzcan E, Isisag A, Sen S, et al. Reproducibility of the 1998 World Health Organization/International Society of Urologic Pathology classification of papillary urothelial neoplasms of the urinary bladder. Virchows Arch. 2003;443(6):734-40.

https://doi.org/10.1007/s00428-003-0905-0

Gonul, II, Poyraz A, Unsal C, Acar C, Alkibay T. Comparison of 1998 WHO/ISUP and 1973 WHO classifications for interobserver variability in grading of papillary urothelial neoplasms of the bladder. Pathological evaluation of 258 cases. Urol Int. 2007;78(4):338-44.

https://doi.org/10.1159/000100839

Mangrud OM, Waalen R, Gudlaugsson E, Dalen I, Tasdemir I, Janssen EA, et al. Reproducibility and prognostic value of WHO1973 and WHO2004 grading systems in TaT1 urothelial carcinoma of the urinary bladder. PLoS One. 2014;9(1):e83192.

https://doi.org/10.1371/journal.pone.0083192

Lopez-Beltran A, Henriques V, Montironi R, Cimadamore A, Raspollini MR, Cheng L. Variants and new entities of bladder cancer. Histopathology. 2019;74(1):77-96.

https://doi.org/10.1111/his.13752

Wang G, McKenney JK. Urinary Bladder Pathology: World Health Organization Classification and American Joint Committee on Cancer Staging Update. Arch Pathol Lab Med. 2019;143(5):571-7.

https://doi.org/10.5858/arpa.2017-0539-RA

Williamson SR, Zhang S, Lopez-Beltran A, Shah RB, Montironi R, Tan PH, et al. Lymphoepithelioma-like carcinoma of the urinary bladder: clinicopathologic, immunohistochemical, and molecular features. Am J Surg Pathol. 2011;35(4):474-83.

https://doi.org/10.1097/PAS.0b013e31820f709e

Willis DL, Fernandez MI, Dickstein RJ, Parikh S, Shah JB, Pisters LL, et al. Clinical outcomes of cT1 micropapillary bladder cancer. J Urol. 2015;193(4):1129-34.

https://doi.org/10.1016/j.juro.2014.09.092

Ikegami H, Iwasaki H, Ohjimi Y, Takeuchi T, Ariyoshi A, Kikuchi M. Sarcomatoid carcinoma of the urinary bladder: a clinicopathologic and immunohistochemical analysis of 14 patients. Hum Pathol. 2000;31(3):332-40.

https://doi.org/10.1016/S0046-8177(00)80247-6

Lawrence MS, Stojanov P, Polak P, Kryukov GV, Cibulskis K, Sivachenko A, et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature. 2013;499(7457):214-8.

https://doi.org/10.1038/nature12213

Network CGAR. Comprehensive molecular characterization of urothelial bladder carcinoma. Nature. 2014;507(7492):315-22.

https://doi.org/10.1038/nature12965

Obermann EC, Meyer S, Hellge D, Zaak D, Filbeck T, Stoehr R, et al. Fluorescence in situ hybridization detects frequent chromosome 9 deletions and aneuploidy in histologically normal urothelium of bladder cancer patients. Oncol Rep. 2004;11(4):745-51.

https://doi.org/10.3892/or.11.4.745

Chow NH, Cairns P, Eisenberger CF, Schoenberg MP, Taylor DC, Epstein JI, et al. Papillary urothelial hyperplasia is a clonal precursor to papillary transitional cell bladder cancer. Int J Cancer. 2000;89(6):514-8.

https://doi.org/10.1002/1097-0215(20001120)89:6<514::AID-IJC8>3.0.CO;2-H

McConkey DJ, Lee S, Choi W, Tran M, Majewski T, Lee S, et al. Molecular genetics of bladder cancer: Emerging mechanisms of tumor initiation and progression. Urol Oncol. 2010;28(4):429-40.

https://doi.org/10.1016/j.urolonc.2010.04.008

Bertz S, Eckstein M, Stoehr R, Weyerer V, Hartmann A. Urothelial Bladder Cancer: An Update on Molecular Pathology with Clinical Implications. Eur Urol Suppl. 2017;16(12):272-94.

https://doi.org/10.1016/j.eursup.2017.10.003

Hernández S, López-Knowles E, Lloreta J, Kogevinas M, Amorós A, Tardón A, et al. Prospective study of FGFR3 mutations as a prognostic factor in nonmuscle invasive urothelial bladder carcinomas. J Clin Oncol. 2006;24(22):3664-71.

https://doi.org/10.1200/JCO.2005.05.1771

López-Knowles E, Hernández S, Malats N, Kogevinas M, Lloreta J, Carrato A, et al. PIK3CA mutations are an early genetic alteration associated with FGFR3 mutations in superficial papillary bladder tumors. Cancer Res. 2006;66(15):7401-4.

https://doi.org/10.1158/0008-5472.CAN-06-1182

Hurst CD, Alder O, Platt FM, Droop A, Stead LF, Burns JE, et al. Genomic Subtypes of Non-invasive Bladder Cancer with Distinct Metabolic Profile and Female Gender Bias in KDM6A Mutation Frequency. Cancer Cell. 2017;32(5):701-15.e7.

https://doi.org/10.1016/j.ccell.2017.08.005

Robertson AG, Kim J, Al-Ahmadie H, Bellmunt J, Guo G, Cherniack AD, et al. Comprehensive Molecular Characterization of Muscle-Invasive Bladder Cancer. Cell. 2017;171(3):540-56.e25.

Knowles MA, Hurst CD. Molecular biology of bladder cancer: new insights into pathogenesis and clinical diversity. Nat Rev Cancer. 2015;15(1):25-41.

https://doi.org/10.1038/nrc3817

Cappellen D, Gil Diez de Medina S, Chopin D, Thiery JP, Radvanyi F. Frequent loss of heterozygosity on chromosome 10q in muscle-invasive transitional cell carcinomas of the bladder. Oncogene. 1997;14(25):3059-66.

https://doi.org/10.1038/sj.onc.1201154

Gildea JJ, Herlevsen M, Harding MA, Gulding KM, Moskaluk CA, Frierson HF, et al. PTEN can inhibit in vitro organotypic and in vivo orthotopic invasion of human bladder cancer cells even in the absence of its lipid phosphatase activity. Oncogene. 2004;23(40):6788-97.

https://doi.org/10.1038/sj.onc.1207599

Allory Y, Beukers W, Sagrera A, Flández M, Marqués M, Márquez M, et al. Telomerase reverse transcriptase promoter mutations in bladder cancer: high frequency across stages, detection in urine, and lack of association with outcome. Eur Urol. 2014;65(2):360-6.

https://doi.org/10.1016/j.eururo.2013.08.052

Kompier LC, Lurkin I, van der Aa MN, van Rhijn BW, van der Kwast TH, Zwarthoff EC. FGFR3, HRAS, KRAS, NRAS and PIK3CA mutations in bladder cancer and their potential as biomarkers for surveillance and therapy. PLoS One. 2010;5(11):e13821.

https://doi.org/10.1371/journal.pone.0013821

Jarvis MC, Ebrahimi D, Temiz NA, Harris RS. Mutation Signatures Including APOBEC in Cancer Cell Lines. JNCI cancer spectrum. 2018;2(1).

https://doi.org/10.1093/jncics/pky002

Roberts SA, Lawrence MS, Klimczak LJ, Grimm SA, Fargo D, Stojanov P, et al. An APOBEC cytidine deaminase mutagenesis pattern is widespread in human cancers. Nat Genet. 2013;45(9):970-6.

https://doi.org/10.1038/ng.2702

Shi MJ, Meng XY, Fontugne J, Chen CL, Radvanyi F, Bernard- Pierrot I. Identification of new driver and passenger mutations within APOBEC-induced hotspot mutations in bladder cancer. Genome Med. 2020;12(1):85.

https://doi.org/10.1186/s13073-020-00781-y

Shi MJ, Meng XY, Lamy P, Banday AR, Yang J, Moreno-Vega A, et al. APOBEC-mediated Mutagenesis as a Likely Cause of FGFR3 S249C Mutation Over-representation in Bladder Cancer. Eur Urol. 2019;76(1):9-13.

https://doi.org/10.1016/j.eururo.2019.03.032

Hedegaard J, Lamy P, Nordentoft I, Algaba F, Høyer S, Ulhøi BP, et al. Comprehensive Transcriptional Analysis of Early-Stage Urothelial Carcinoma. Cancer Cell. 2016;30(1):27-42.

https://doi.org/10.1016/j.ccell.2016.05.004

Sjödahl G, Lauss M, Lövgren K, Chebil G, Gudjonsson S, Veerla S, et al. A molecular taxonomy for urothelial carcinoma. Clin Cancer Res. 2012;18(12):3377-86.

https://doi.org/10.1158/1078-0432.CCR-12-0077-T

Sjödahl G, Eriksson P, Liedberg F, Höglund M. Molecular classification of urothelial carcinoma: global mRNA classification versus tumour-cell phenotype classification. J Pathol. 2017;242(1):113- 25.

https://doi.org/10.1002/path.4886

Damrauer JS, Hoadley KA, Chism DD, Fan C, Tiganelli CJ, Wobker SE, et al. Intrinsic subtypes of high-grade bladder cancer reflect the hallmarks of breast cancer biology. Proc Natl Acad Sci U S A. 2014;111(8):3110-5.

https://doi.org/10.1073/pnas.1318376111

Perou CM, Sørlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, et al. Molecular portraits of human breast tumours. Nature. 2000;406(6797):747-52.

https://doi.org/10.1038/35021093

Prat A, Karginova O, Parker JS, Fan C, He X, Bixby L, et al. Characterization of cell lines derived from breast cancers and normal mammary tissues for the study of the intrinsic molecular subtypes. Breast Cancer Res Treat. 2013;142(2):237-55.

https://doi.org/10.1007/s10549-013-2743-3

Kardos J, Chai S, Mose LE, Selitsky SR, Krishnan B, Saito R, et al. Claudin-low bladder tumors are immune infiltrated and actively immune suppressed. JCI insight. 2016;1(3):e85902.

https://doi.org/10.1172/jci.insight.85902

Choi W, Porten S, Kim S, Willis D, Plimack ER, Hoffman- Censits J, et al. Identification of distinct basal and luminal subtypes of muscle-invasive bladder cancer with different sensitivities to frontline chemotherapy. Cancer Cell. 2014;25(2):152-65.

https://doi.org/10.1016/j.ccr.2014.01.009

Dadhania V, Zhang M, Zhang L, Bondaruk J, Majewski T, Siefker-Radtke A, et al. Meta-Analysis of the Luminal and Basal Subtypes of Bladder Cancer and the Identification of Signature Immunohistochemical Markers for Clinical Use. EBioMedicine. 2016;12:105-17.

https://doi.org/10.1016/j.ebiom.2016.08.036

Aine M, Eriksson P, Liedberg F, Höglund M, Sjödahl G. On Molecular Classification of Bladder Cancer: Out of One, Many. Eur Urol. 2015;68(6):921-3.

https://doi.org/10.1016/j.eururo.2015.07.021

Kamoun A, de Reyniès A, Allory Y, Sjödahl G, Robertson AG, Seiler R, et al. A Consensus Molecular Classification of Muscle-invasive Bladder Cancer. Eur Urol. 2020;77(4):420-33.

https://doi.org/10.1016/j.eururo.2019.09.006

AJCC Cancer Staging Manual. Eight Edition ed: Springer; 2017.

Miller KD, Nogueira L, Mariotto AB, Rowland JH, Yabroff KR, Alfano CM, et al. Cancer treatment and survivorship statistics, 2019. CA Cancer J Clin. 2019;69(5):363-85.

https://doi.org/10.3322/caac.21565

Bol MG, Baak JP, Buhr-Wildhagen S, Kruse AJ, Kjellevold KH, Janssen EA, et al. Reproducibility and prognostic variability of grade and lamina propria invasion in stages Ta, T1 urothelial carcinoma of the bladder. J Urol. 2003;169(4):1291-4.

https://doi.org/10.1097/01.ju.0000055471.78783.ae

van Rhijn BW, van der Kwast TH, Alkhateeb SS, Fleshner NE, van Leenders GJ, Bostrom PJ, et al. A new and highly prognostic system to discern T1 bladder cancer substage. Eur Urol. 2012;61(2):378-84.

https://doi.org/10.1016/j.eururo.2011.10.026

Magers MJ, Lopez-Beltran A, Montironi R, Williamson SR, Kaimakliotis HZ, Cheng L. Staging of bladder cancer. Histopathology. 2019;74(1):112-34.

https://doi.org/10.1111/his.13734

Sylvester RJ, van der Meijden AP, Oosterlinck W, Witjes JA, Bouffioux C, Denis L, et al. Predicting recurrence and progression in individual patients with stage Ta T1 bladder cancer using EORTC risk tables: a combined analysis of 2596 patients from seven EORTC trials. Eur Urol. 2006;49(3):466-5; discussion 75-7.

https://doi.org/10.1016/j.eururo.2005.12.031

Kim HS, Kim M, Jeong CW, Kwak C, Kim HH, Ku JH. Presence of lymphovascular invasion in urothelial bladder cancer specimens after transurethral resections correlates with risk of upstaging and survival: a systematic review and meta-analysis. Urol Oncol. 2014;32(8):1191-9.

https://doi.org/10.1016/j.urolonc.2014.05.008

Bol MG, Baak JP, de Bruin PC, Rep S, Marx W, Bos S, et al. Improved objectivity of grading of T(A,1) transitional cell carcinomas of the urinary bladder by quantitative nuclear and proliferation related features. J Clin Pathol. 2001;54(11):854-9.

https://doi.org/10.1136/jcp.54.11.854

Bol MG, Baak JP, Rep S, Marx WL, Kruse AJ, Bos SD, et al. Prognostic value of proliferative activity and nuclear morphometry for progression in TaT1 urothelial cell carcinomas of the urinary bladder. Urology. 2002;60(6):1124-30.

https://doi.org/10.1016/S0090-4295(02)01906-4

van Rhijn BW, Vis AN, van der Kwast TH, Kirkels WJ, Radvanyi F, Ooms EC, et al. Molecular grading of urothelial cell carcinoma with fibroblast growth factor receptor 3 and MIB-1 is superior to pathologic grade for the prediction of clinical outcome. J Clin Oncol. 2003;21(10):1912-21.

https://doi.org/10.1200/JCO.2003.05.073

Bertz S, Otto W, Denzinger S, Wieland WF, Burger M, Stöhr R, et al. Combination of CK20 and Ki-67 immunostaining analysis predicts recurrence, progression, and cancer-specific survival in pT1 urothelial bladder cancer. Eur Urol. 2014;65(1):218-26.

https://doi.org/10.1016/j.eururo.2012.05.033

Mangrud OM, Gudlaugsson E, Skaland I, Tasdemir I, Dalen I, van Diermen B, et al. Prognostic comparison of proliferation markers and World Health Organization 1973/2004 grades in urothelial carcinomas of the urinary bladder. Hum Pathol. 2014;45(7):1496-503.

https://doi.org/10.1016/j.humpath.2014.03.001

Amin MB, Trpkov K, Lopez-Beltran A, Grignon D. Best practices recommendations in the application of immunohistochemistry in the bladder lesions: report from the International Society of Urologic Pathology consensus conference. Am J Surg Pathol. 2014;38(8):e20-34.

https://doi.org/10.1097/PAS.0000000000000240

Harnden P, Eardley I, Joyce AD, Southgate J. Cytokeratin 20 as an objective marker of urothelial dysplasia. Br J Urol. 1996;78(6):870- 5.

https://doi.org/10.1046/j.1464-410X.1996.23511.x

Alston ELJ, Zynger DL. Does the addition of AMACR to CK20 help to diagnose challenging cases of urothelial carcinoma in situ? Diagn Pathol. 2019;14(1):91.

https://doi.org/10.1186/s13000-019-0871-8

Desai S, Lim SD, Jimenez RE, Chun T, Keane TE, McKenney JK, et al. Relationship of cytokeratin 20 and CD44 protein expression with WHO/ISUP grade in pTa and pT1 papillary urothelial neoplasia. Mod Pathol. 2000;13(12):1315-23.

https://doi.org/10.1038/modpathol.3880241

Harnden P, Mahmood N, Southgate J. Expression of cytokeratin20 redefines urothelial papillomas of the bladder. Lancet. 1999;353(9157):974-7.

https://doi.org/10.1016/S0140-6736(98)05383-5

Esrig D, Spruck CH, 3rd, Nichols PW, Chaiwun B, Steven K, Groshen S, et al. p53 nuclear protein accumulation correlates with mutations in the p53 gene, tumor grade, and stage in bladder cancer. Am J Pathol. 1993;143(5):1389-97.

Kelsey KT, Hirao T, Schned A, Hirao S, Devi-Ashok T, Nelson HH, et al. A population-based study of immunohistochemical detection of p53 alteration in bladder cancer. Br J Cancer. 2004;90(8):1572-6.

https://doi.org/10.1038/sj.bjc.6601748

Hodgson A, Xu B, Downes MR. p53 immunohistochemistry in high-grade urothelial carcinoma of the bladder is prognostically significant. Histopathology. 2017;71(2):296-304.

https://doi.org/10.1111/his.13225

Esrig D, Elmajian D, Groshen S, Freeman JA, Stein JP, Chen SC, et al. Accumulation of nuclear p53 and tumor progression in bladder cancer. N Engl J Med. 1994;331(19):1259-64.

https://doi.org/10.1056/NEJM199411103311903

Gooden MJ, de Bock GH, Leffers N, Daemen T, Nijman HW. The prognostic influence of tumour-infiltrating lymphocytes in cancer: a systematic review with meta-analysis. Br J Cancer. 2011;105(1):93-103.

https://doi.org/10.1038/bjc.2011.189

Mahmoud SM, Paish EC, Powe DG, Macmillan RD, Grainge MJ, Lee AH, et al. Tumor-infiltrating CD8+ lymphocytes predict clinical outcome in breast cancer. J Clin Oncol. 2011;29(15):1949-55.

https://doi.org/10.1200/JCO.2010.30.5037

Ooft ML, van Ipenburg JA, Braunius WW, Zuur CI, Koljenović S, Willems SM. Prognostic role of tumor infiltrating lymphocytes in EBV positive and EBV negative nasopharyngeal carcinoma. Oral Oncol. 2017;71:16-25.

https://doi.org/10.1016/j.oraloncology.2017.05.015

Liu K, Zhao K, Wang L, Sun E. The prognostic values of tumor- infiltrating neutrophils, lymphocytes and neutrophil/lymphocyte rates in bladder urothelial cancer. Pathol Res Pract. 2018;214(8):1074-80.

https://doi.org/10.1016/j.prp.2018.05.010

Sharma P, Shen Y, Wen S, Yamada S, Jungbluth AA, Gnjatic S, et al. CD8 tumor-infiltrating lymphocytes are predictive of survival in muscle-invasive urothelial carcinoma. Proc Natl Acad Sci U S A. 2007;104(10):3967-72.

https://doi.org/10.1073/pnas.0611618104

Yu A, Mansure JJ, Solanki S, Siemens DR, Koti M, Dias ABT, et al. Presence of lymphocytic infiltrate cytotoxic T lymphocyte CD3+, CD8+, and immunoscore as prognostic marker in patients after radical cystectomy. PLoS One. 2018;13(10):e0205746.

https://doi.org/10.1371/journal.pone.0205746

Faraj SF, Munari E, Guner G, Taube J, Anders R, Hicks J, et al. Assessment of tumoral PD-L1 expression and intratumoral CD8+ T cells in urothelial carcinoma. Urology. 2015;85(3):703.e1-6.

https://doi.org/10.1016/j.urology.2014.10.020

Wahlin S, Nodin B, Leandersson K, Boman K, Jirström K. Clinical impact of T cells, B cells and the PD-1/PD-L1 pathway in muscle invasive bladder cancer: a comparative study of transurethral resection and cystectomy specimens. Oncoimmunology. 2019;8(11):e1644108.

https://doi.org/10.1080/2162402X.2019.1644108

Zhang S, Wang J, Zhang X, Zhou F. Tumor-infiltrating CD8+ lymphocytes predict different clinical outcomes in organ- and non- organ-confined urothelial carcinoma of the bladder following radical cystectomy. PeerJ. 2017;5:e3921.

https://doi.org/10.7717/peerj.3921

Horn T, Laus J, Seitz AK, Maurer T, Schmid SC, Wolf P, et al. The prognostic effect of tumour-infiltrating lymphocytic subpopulations in bladder cancer. World J Urol. 2016;34(2):181-7.

https://doi.org/10.1007/s00345-015-1615-3

Zhang Q, Hao C, Cheng G, Wang L, Wang X, Li C, et al. High CD4⁺ T cell density is associated with poor prognosis in patients with non-muscle-invasive bladder cancer. Int J Clin Exp Pathol. 2015;8(9):11510-6.

Yano H, Andrews LP, Workman CJ, Vignali DAA. Intratumoral regulatory T cells: markers, subsets and their impact on anti-tumor immunity. Immunology. 2019;157(3):232-47.

https://doi.org/10.1111/imm.13067

Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med. 2004;10(9):942-9.

https://doi.org/10.1038/nm1093

Miyake M, Tatsumi Y, Gotoh D, Ohnishi S, Owari T, Iida K, et al. Regulatory T Cells and Tumor-Associated Macrophages in the Tumor Microenvironment in Non-Muscle Invasive Bladder Cancer Treated with Intravesical Bacille Calmette-Guérin: A Long-Term Follow-Up Study of a Japanese Cohort. International journal of molecular sciences. 2017;18(10).

https://doi.org/10.3390/ijms18102186

Winerdal ME, Marits P, Winerdal M, Hasan M, Rosenblatt R, Tolf A, et al. FOXP3 and survival in urinary bladder cancer. BJU Int. 2011;108(10):1672-8.

https://doi.org/10.1111/j.1464-410X.2010.10020.x

Parodi A, Traverso P, Kalli F, Conteduca G, Tardito S, Curto M, et al. Residual tumor micro-foci and overwhelming regulatory T lymphocyte infiltration are the causes of bladder cancer recurrence. Oncotarget. 2016;7(6):6424-35.

https://doi.org/10.18632/oncotarget.7024

Miyake M, Hori S, Owari T, Oda Y, Tatsumi Y, Nakai Y, et al. Clinical Impact of Tumor-Infiltrating Lymphocytes and PD-L1-Positive Cells as Prognostic and Predictive Biomarkers in Urological Malignancies and Retroperitoneal Sarcoma. Cancers (Basel). 2020;12(11).

https://doi.org/10.3390/cancers12113153

Brauers A, Buettner R, Jakse G. Second resection and prognosis of primary high risk superficial bladder cancer: is cystectomy often too early? J Urol. 2001;165(3):808-10.

https://doi.org/10.1016/S0022-5347(05)66532-9

https://doi.org/10.1097/00005392-200103000-00017

Sylvester RJ, Oosterlinck W, van der Meijden AP. A single immediate postoperative instillation of chemotherapy decreases the risk of recurrence in patients with stage Ta T1 bladder cancer: a meta- analysis of published results of randomized clinical trials. J Urol. 2004;171(6 Pt 1):2186-90, quiz 435.

https://doi.org/10.1097/01.ju.0000125486.92260.b2

Morales A, Eidinger D, Bruce AW. Intracavitary Bacillus Calmette-Guerin in the treatment of superficial bladder tumors. J Urol. 1976;116(2):180-3.

https://doi.org/10.1016/S0022-5347(17)58737-6

Oddens J, Brausi M, Sylvester R, Bono A, van de Beek C, van Andel G, et al. Final results of an EORTC-GU cancers group randomized study of maintenance bacillus Calmette-Guérin in intermediate- and high-risk Ta, T1 papillary carcinoma of the urinary bladder: one-third dose versus full dose and 1 year versus 3 years of maintenance. Eur Urol. 2013;63(3):462-72.

https://doi.org/10.1016/j.eururo.2012.10.039

Malmström PU, Sylvester RJ, Crawford DE, Friedrich M, Krege S, Rintala E, et al. An individual patient data meta-analysis of the long- term outcome of randomised studies comparing intravesical mitomycin C versus bacillus Calmette-Guérin for non-muscle-invasive bladder cancer. Eur Urol. 2009;56(2):247-56.

https://doi.org/10.1016/j.eururo.2009.04.038

Han RF, Pan JG. Can intravesical bacillus Calmette-Guérin reduce recurrence in patients with superficial bladder cancer? A meta- analysis of randomized trials. Urology. 2006;67(6):1216-23.

https://doi.org/10.1016/j.urology.2005.12.014

Böhle A, Bock PR. Intravesical bacille Calmette-Guérin versus mitomycin C in superficial bladder cancer: formal meta-analysis of comparative studies on tumor progression. Urology. 2004;63(4):682-6; discussion 6-7.

https://doi.org/10.1016/j.urology.2003.11.049

Sylvester RJ, van der MA, Lamm DL. Intravesical bacillus Calmette-Guerin reduces the risk of progression in patients with superficial bladder cancer: a meta-analysis of the published results of randomized clinical trials. J Urol. 2002;168(5):1964-70.

https://doi.org/10.1016/S0022-5347(05)64273-5

https://doi.org/10.1097/00005392-200211000-00016

Boehm BE, Cornell JE, Wang H, Mukherjee N, Oppenheimer JS, Svatek RS. Efficacy of bacillus Calmette-Guérin Strains for Treatment of Nonmuscle Invasive Bladder Cancer: A Systematic Review and Network Meta-Analysis. J Urol. 2017;198(3):503-10.

https://doi.org/10.1016/j.juro.2017.01.086

Larsen ES, Joensen UN, Poulsen AM, Goletti D, Johansen IS. Bacillus Calmette-Guérin immunotherapy for bladder cancer: a review of immunological aspects, clinical effects and BCG infections. APMIS. 2020;128(2):92-103.

https://doi.org/10.1111/apm.13011

Kawai K, Miyazaki J, Joraku A, Nishiyama H, Akaza H. Bacillus Calmette-Guerin (BCG) immunotherapy for bladder cancer: current understanding and perspectives on engineered BCG vaccine. Cancer Sci. 2013;104(1):22-7.

https://doi.org/10.1111/j.1349-7006.2009.01373.x

https://doi.org/10.1111/cas.12075

Bevers RF, de Boer EC, Kurth KH, Schamhart DH. BCG- induced interleukin-6 upregulation and BCG internalization in well and poorly differentiated human bladder cancer cell lines. Eur Cytokine Netw. 1998;9(2):181-6.

Bisiaux A, Thiounn N, Timsit MO, Eladaoui A, Chang HH, Mapes J, et al. Molecular analyte profiling of the early events and tissue conditioning following intravesical bacillus calmette-guerin therapy in patients with superficial bladder cancer. J Urol. 2009;181(4):1571-80.

https://doi.org/10.1016/j.juro.2008.11.124

Luo Y. Blocking IL-10 enhances bacillus Calmette-Guérin induced T helper Type 1 immune responses and anti-bladder cancer immunity. Oncoimmunology. 2012;1(7):1183-5.

https://doi.org/10.4161/onci.20640

Ratliff TL, Ritchey JK, Yuan JJ, Andriole GL, Catalona WJ. T- cell subsets required for intravesical BCG immunotherapy for bladder cancer. J Urol. 1993;150(3):1018-23.

https://doi.org/10.1016/S0022-5347(17)35678-1

Pichler R, Fritz J, Zavadil C, Schäfer G, Culig Z, Brunner A. Tumor-infiltrating immune cell subpopulations influence the oncologic outcome after intravesical Bacillus Calmette-Guérin therapy in bladder cancer. Oncotarget. 2016;7(26):39916-30.

https://doi.org/10.18632/oncotarget.9537

Brausi M, Oddens J, Sylvester R, Bono A, van de Beek C, van Andel G, et al. Side effects of Bacillus Calmette-Guérin (BCG) in the treatment of intermediate- and high-risk Ta, T1 papillary carcinoma of the bladder: results of the EORTC genito-urinary cancers group randomised phase 3 study comparing one-third dose with full dose and 1 year with 3 years of maintenance BCG. Eur Urol. 2014;65(1):69-76.

https://doi.org/10.1016/j.eururo.2013.07.021

Lamm DL, Blumenstein BA, Crissman JD, Montie JE, Gottesman JE, Lowe BA, et al. Maintenance bacillus Calmette-Guerin immunotherapy for recurrent TA, T1 and carcinoma in situ transitional cell carcinoma of the bladder: a randomized Southwest Oncology Group Study. J Urol. 2000;163(4):1124-9.

https://doi.org/10.1016/S0022-5347(05)67707-5

https://doi.org/10.1097/00005392-200004000-00014

Larsen ES, Nordholm AC, Lillebaek T, Holden IK, Johansen IS. The epidemiology of bacille Calmette-Guérin infections after bladder instillation from 2002 through 2017: a nationwide retrospective cohort study. BJU Int. 2019;124(6):910-6.

https://doi.org/10.1111/bju.14793

van der Meijden AP, Sylvester RJ, Oosterlinck W, Hoeltl W, Bono AV. Maintenance Bacillus Calmette-Guerin for Ta T1 bladder tumors is not associated with increased toxicity: results from a European Organisation for Research and Treatment of Cancer Genito-Urinary Group Phase III Trial. Eur Urol. 2003;44(4):429-34.

https://doi.org/10.1016/S0302-2838(03)00357-9

Holmang S, Johansson SL. Stage Ta-T1 bladder cancer: the relationship between findings at first followup cystoscopy and subsequent recurrence and progression. J Urol. 2002;167(4):1634-7.

https://doi.org/10.1016/S0022-5347(05)65168-3

https://doi.org/10.1097/00005392-200204000-00014

Fedchenko N, Reifenrath J. Different approaches for interpretation and reporting of immunohistochemistry analysis results in the bone tissue - a review. Diagn Pathol. 2014;9:221.

https://doi.org/10.1186/s13000-014-0221-9

Remmele W, Stegner HE. [Recommendation for uniform definition of an immunoreactive score (IRS) for immunohistochemical estrogen receptor detection (ER-ICA) in breast cancer tissue]. Pathologe. 1987;8(3):138-40.

Gwet KL. Handbook of inter-rater reliability: The definitive guide to measuring the extent of agreement among raters: Advanced Analytics, LLC; 2014.

DG A. Practical statistics for medical research. London: Chapman and Hall/CRC; 1991.

Hassler MR, Bray F, Catto JWF, Grollman AP, Hartmann A, Margulis V, et al. Molecular Characterization of Upper Tract Urothelial Carcinoma in the Era of Next-generation Sequencing: A Systematic Review of the Current Literature. Eur Urol. 2020;78(2):209-20.

https://doi.org/10.1016/j.eururo.2020.05.039

Swartz MA, Porter MP, Lin DW, Weiss NS. Incidence of primary urethral carcinoma in the United States. Urology. 2006;68(6):1164-8.

https://doi.org/10.1016/j.urology.2006.08.1057

van Kessel KEM, van der Keur KA, Dyrskjøt L, Algaba F, Welvaart NYC, Beukers W, et al. Molecular Markers Increase Precision of the European Association of Urology Non-Muscle-Invasive Bladder Cancer Progression Risk Groups. Clin Cancer Res. 2018;24(7):1586-93.

https://doi.org/10.1158/1078-0432.CCR-17-2719

Gontero P, Sylvester R, Pisano F, Joniau S, Oderda M, Serretta V, et al. The impact of re-transurethral resection on clinical outcomes in a large multicentre cohort of patients with T1 high-grade/Grade 3 bladder cancer treated with bacille Calmette-Guérin. BJU Int. 2016;118(1):44- 52.

https://doi.org/10.1111/bju.13354

Griffin J, Treanor D. Digital pathology in clinical use: where are we now and what is holding us back? Histopathology. 2017;70(1):134- 45.

https://doi.org/10.1111/his.12993

Pallua JD, Brunner A, Zelger B, Schirmer M, Haybaeck J. The future of pathology is digital. Pathol Res Pract. 2020;216(9):153040.

https://doi.org/10.1016/j.prp.2020.153040

Niazi MKK, Parwani AV, Gurcan MN. Digital pathology and artificial intelligence. Lancet Oncol. 2019;20(5):e253-e61.

https://doi.org/10.1016/S1470-2045(19)30154-8

Yin PN, Kc K, Wei S, Yu Q, Li R, Haake AR, et al. Histopathological distinction of non-invasive and invasive bladder cancers using machine learning approaches. BMC Med Inform Decis Mak. 2020;20(1):162.

https://doi.org/10.1186/s12911-020-01185-z

Jansen I, Lucas M, Bosschieter J, de Boer OJ, Meijer SL, van Leeuwen TG, et al. Automated Detection and Grading of Non-Muscle- Invasive Urothelial Cell Carcinoma of the Bladder. Am J Pathol. 2020;190(7):1483-90.

https://doi.org/10.1016/j.ajpath.2020.03.013

Cover image

Downloads

Published

March 30, 2022

License

Creative Commons License

This work is licensed under a Creative Commons Attribution 4.0 International License.