Skip to main content

Genitourinary Pathology (Including Adrenal Gland)

  • Chapter
  • First Online:
Oncological Surgical Pathology

Abstract

Our aims in constructing the Genitourinary Pathology chapter are to describe neoplasms of the adrenal gland, urothelial tract, kidney, penis, prostate, and testis in a manner that is both useful for the practicing surgical pathologist and that may be used as a reference for all students of urologic pathology. Whereas the text and figures describe the salient morphologic, immunohistochemical, and molecular attributes for each tumor type and encompass the latest classification schemes, the narrative integrates the clinical and pathological findings that are commonly encountered during surgical pathology sign-out of these cases. Accordingly, it is our hope that this chapter will serve as a guide for both general and subspecialized pathologists in contemporary practice.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 219.00
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
Hardcover Book
USD 279.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Bostwick DG, Cheng L. Precursors of prostate cancer. Histopathology. 2012;60:4ā€“27.

    PubMedĀ  Google ScholarĀ 

  2. Iczkowski KA, Torkko KC, Wilson RS, et al. Prostatic atrophy: its spatial proximity to carcinoma and intraepithelial neoplasia based on annotation of digital slides. Hum Pathol. 2014;45:54ā€“8.

    PubMedĀ  Google ScholarĀ 

  3. Davidson D, Bostwick DG, Qian J, et al. Prostatic intraepithelial neoplasia is a risk factor for adenocarcinoma: predictive accuracy in needle biopsies. J Urol. 1995;154:1295ā€“9.

    PubMedĀ  CASĀ  Google ScholarĀ 

  4. Schlesinger C, Bostwick DG, Iczkowski KA. High-grade prostatic intraepithelial neoplasia and atypical small acinar proliferation: their predictive values for cancer in current practice. Am J Surg Pathol. 2005;29:1201ā€“7.

    PubMedĀ  Google ScholarĀ 

  5. Patel P, Nayak JG, Biljetina Z, et al. Prostate cancer after initial high-grade prostatic intraepithelial neoplasia and benign prostate biopsy. Can J Urol. 2015;22:8056ā€“62.

    PubMedĀ  Google ScholarĀ 

  6. Oderda M, Gontero P. High-grade prostatic intraepithelial neoplasia and atypical small acinar proliferation: is repeat biopsy still necessary? BJU Int. 2009;104:1554ā€“6.

    PubMedĀ  Google ScholarĀ 

  7. Cheng L, Shan A, Cheville JC, Qian J, et al. Atypical adenomatous hyperplasia of the prostate: a premalignant lesion? Cancer Res. 1998;58:389ā€“91.

    PubMedĀ  CASĀ  Google ScholarĀ 

  8. Zhang C, Montironi R, MacLennan GT, et al. Is atypical adenomatous hyperplasia of the prostate a precursor lesion? Prostate. 2011;71:1746ā€“51.

    PubMedĀ  CASĀ  Google ScholarĀ 

  9. De Marzo AM, Marchi VL, Epstein JI, et al. Proliferative inflammatory atrophy of the prostate: implications for prostatic carcinogenesis. Am J Pathol. 1999;155:1985ā€“92.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  10. Servian P, Celma A, Planas J, et al. Clinical significance of proliferative inflammatory atrophy in negative prostatic biopsies. Prostate. 2016;76:1501ā€“6.

    PubMedĀ  Google ScholarĀ 

  11. Renshaw AA. Correlation of gross morphologic features with histologic features in radical prostatectomy specimens. Am J Clin Pathol. 1998;110:38ā€“42.

    PubMedĀ  CASĀ  Google ScholarĀ 

  12. Totten RS, Heinemann MW, Hudson PB, et al. Microscopic differential diagnosis of latent carcinoma of prostate. AMA Arch Pathol. 1953;55:131ā€“41.

    PubMedĀ  CASĀ  Google ScholarĀ 

  13. Iczkowski KA. Prostate pointers and pitfalls: the 10 most prevalent problems in prostate biopsy interpretation. Ann Diagn Pathol. 2014;18:301ā€“11.

    PubMedĀ  Google ScholarĀ 

  14. Iczkowski KA, Bostwick DG. Criteria for biopsy diagnosis of minimal volume prostatic adenocarcinoma: analytic comparison with nondiagnostic but suspicious, atypical small acinar proliferation. Arch Pathol Lab Med. 2000;124:98ā€“107.

    PubMedĀ  CASĀ  Google ScholarĀ 

  15. Algaba F, Epstein JI, Aldape HC, et al. Assessment of prostate carcinoma in core needle biopsy --definition of minimal criteria for the diagnosis of cancer in biopsy material. Cancer. 1996;78:376ā€“81.

    PubMedĀ  CASĀ  Google ScholarĀ 

  16. Baisden BL, Kahane H, Epstein JI. Perineural invasion, mucinous fibroplasia, and glomerulations; diagnostic features of limited cancer on prostate needle biopsy. Am J Surg Pathol. 1999;23:918ā€“24.

    PubMedĀ  CASĀ  Google ScholarĀ 

  17. Arangelovich V, Tretiakova M, SenGupta E, et al. Pathogenesis and significance of collagenous micronodules of the prostate. Appl Immunohistochem Mol Morphol. 2003;11:15ā€“9.

    PubMedĀ  Google ScholarĀ 

  18. Niroomand H, Nowroozi M, Ayati M, et al. Relationship between perineural invasion in prostate needle biopsy specimens and pathologic staging after radical prostatectomy. Nephrourol Mon. 2016;8(3):e36022.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  19. Vargas SO, Jiroutek M, Welch WR, et al. Perineural invasion in prostate needle biopsy specimens. Correlation with extraprostatic extension at resection. Am J Clin Pathol. 1999;111:223ā€“8.

    PubMedĀ  CASĀ  Google ScholarĀ 

  20. Haki Yuksel O, Urkmez A, Verit A. Can perineural invasion detected in prostate needle biopsy specimens predict surgical margin positivity in Dā€™Amico low risk patients? Arch Ital Urol Androl. 2016;88:89ā€“92.

    PubMedĀ  Google ScholarĀ 

  21. Morlacco A, Cheville JC, Rangel LJ, et al. Adverse disease features in Gleason score 3 + 4 ā€œfavorable intermediate-riskā€ prostate cancer: implications for active surveillance. Eur Urol. 2017;72(3):442ā€“47.

    Google ScholarĀ 

  22. Bismar TA, Lewis JS, Vollmer RT, et al. Multiple measures of carcinoma extent versus perineural invasion in prostate needle biopsy tissue in prediction of pathologic stage in a screening population. Am J Surg Pathol. 2003;27:432ā€“40.

    PubMedĀ  Google ScholarĀ 

  23. Shah RB, Leandro G, Romerocaces G, et al. Improvement of diagnostic agreement among pathologists in resolving an ā€œatypical glands suspicious for cancerā€ diagnosis in prostate biopsies utilizing the nodule diseaseĀ ā€“ focused diagnostic review quality improvement process. Hum Pathol. 2016;56:155ā€“62.

    PubMedĀ  Google ScholarĀ 

  24. Iczkowski KA, Cheng L, Crawford BG, et al. Steam heat with an EDTA buffer and protease digestion optimizes immunohistochemical expression of basal cell-specific antikeratin 34betaE12 to discriminate cancer in prostatic epithelium. Mod Pathol. 1999;12:1ā€“4.

    PubMedĀ  CASĀ  Google ScholarĀ 

  25. Abrahams NA, Bostwick DG, Ormsby AH, et al. Distinguishing atrophy and high-grade prostatic intraepithelial neoplasia from prostatic adenocarcinoma with and without previous adjuvant hormone therapy with the aid of cytokeratin 5/6. Am J Clin Pathol. 2003;120:368ā€“76.

    PubMedĀ  Google ScholarĀ 

  26. Jiang Z, Iczkowski KA, Woda BA, et al. P504S immunostaining boosts diagnostic resolution of suspicious foci in prostatic needle biopsies. Am J Clin Pathol. 2004;121:99ā€“107.

    PubMedĀ  Google ScholarĀ 

  27. Goldstein J, Goyal R, Roland JT, et al. MAGI-2 is a sensitive and specific marker of prostatic adenocarcinoma: a comparison with AMACR. Am J Clin Pathol. 2016;146:294ā€“302.

    PubMedĀ  CASĀ  Google ScholarĀ 

  28. Qian J, Hirasawa K, Bostwick DG, et al. Loss of p53 and c-myc overrepresentation in stage T(2-3)N(1-3)M(0) prostate cancer are potential markers for cancer progression. Mod Pathol. 2002;15:35ā€“44.

    PubMedĀ  Google ScholarĀ 

  29. Bostwick DG, Cheng L. Urologic surgical pathology. 3rd ed. St. Louis: Mosby; 2014.

    Google ScholarĀ 

  30. Epstein JI, Egevad L, Humphrey PA, Montironi R, Members of the ISUP Immunohistochemistry in Diagnostic Urologic Pathology Group. Best practices recommendations in the application of immunohistochemistry in the prostate: report from the International Society of Urologic Pathology consensus conference. Am J Surg Pathol. 2014;38:e6ā€“e19.

    PubMedĀ  Google ScholarĀ 

  31. Plourde A, Gross A, Jiang Z, et al. Patterns in immunohistochemical usage in extended core prostate biopsies: comparisons among genitourinary pathologists and nongenitourinary pathologists. Arch Pathol Lab Med. 2013;137:1630ā€“4.

    PubMedĀ  Google ScholarĀ 

  32. Hameed O, Humphrey PA. Immunohistochemistry in diagnostic surgical pathology of the prostate. Semin Diagn Pathol. 2005;22:88ā€“104.

    PubMedĀ  Google ScholarĀ 

  33. Varma M, Jasani B. Diagnostic utility of immunohistochemistry in morphologically difficult prostate cancer: review of current literature. Histopathology. 2005;47:1ā€“16.

    PubMedĀ  CASĀ  Google ScholarĀ 

  34. Humphrey PA, Kaleem Z, Swanson PE, et al. Pseudohyperplastic prostatic adenocarcinoma. Am J Surg Pathol. 1998;22:1239ā€“46.

    PubMedĀ  CASĀ  Google ScholarĀ 

  35. Xiao GQ, Burstein DE, Miller LK, et al. Nephrogenic adenoma: immunohistochemical evaluation for its etiology and differentiation from prostatic adenocarcinoma. Arch Pathol Lab Med. 2006;130:805ā€“10.

    PubMedĀ  Google ScholarĀ 

  36. Iczkowski KA. Paneth cell-like change in benign prostate can account for P504S (AMACR) reactivity. Int J Clin Exp Pathol. 2014;7:3454ā€“5. eCollection 2014.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  37. Srigley JR. Benign mimickers of prostatic adenocarcinoma. Mod Pathol. 2004;17:328ā€“48.

    PubMedĀ  Google ScholarĀ 

  38. Hameed O, Humphrey PA. Pseudoneoplastic mimics of prostate and bladder carcinomas. Arch Pathol Lab Med. 2010;134:427ā€“43.

    PubMedĀ  Google ScholarĀ 

  39. Kaleem Z, Swanson PE, Vollmer RT, et al. Prostatic adenocarcinoma with atrophic features: a study of 202 consecutive completely embedded radical prostatectomy specimens. Am J Clin Pathol. 1998;109:695ā€“703.

    PubMedĀ  CASĀ  Google ScholarĀ 

  40. Yaskiv O, Cao D, Humphrey PA. Microcystic adenocarcinoma of the prostate: a variant of pseudohyperplastic and atrophic patterns. Am J Surg Pathol. 2010;34:556ā€“61.

    PubMedĀ  Google ScholarĀ 

  41. Muezzinoglu B, Erdamar S, et al. Verumontanum mucosal gland hyperplasia is associated with atypical adenomatous hyperplasia of the prostate. Arch Pathol Lab Med. 2001;125:358ā€“60.

    PubMedĀ  CASĀ  Google ScholarĀ 

  42. Knoedler JJ, Karnes RJ, Thompson RH, et al. The association of tumor volume with mortality following radical prostatectomy. Prostate Cancer Prostatic Dis. 2014;17:144ā€“8.

    PubMedĀ  CASĀ  Google ScholarĀ 

  43. Lerner SP, Shen S. Pathologic assessment and clinical significance of prostatic involvement by transitional cell carcinoma and prostate cancer. Urol Oncol. 2008;26:481ā€“5.

    PubMedĀ  Google ScholarĀ 

  44. Revelo MP, Cookson MS, Chang SS, et al. Incidence and location of prostate and urothelial carcinoma in prostates from cystoprostatectomies: implications for possible apical sparing surgery. J Urol. 2008;179(5 Suppl):S27ā€“32.

    PubMedĀ  Google ScholarĀ 

  45. Wood DP, Montie JE, Pontes JE, et al. Transitional cell carcinoma of the prostate in cystoprostatectomy specimens removed for bladder cancer. J Urol. 1989;141:346ā€“9.

    PubMedĀ  Google ScholarĀ 

  46. Mazzucchelli R, Barbisan F, Scarpelli M, et al. Is incidentally detected prostate cancer in patients undergoing radical cystoprostatectomy clinically significant? Am J Clin Pathol. 2009;131:279ā€“83.

    PubMedĀ  Google ScholarĀ 

  47. Smith SC, Mohanty SK, Kunju LP, et al. Uroplakin II outperforms uroplakin III in diagnostically challenging settings. Histopathology. 2014;65:132ā€“8.

    PubMedĀ  Google ScholarĀ 

  48. Tian W, Guner G, Miyamoto H, et al. Utility of Uroplakin II expression as a marker of urothelial carcinoma. Hum Pathol. 2015;46:58ā€“64.

    PubMedĀ  Google ScholarĀ 

  49. Osunkoya AO, Netto GJ, Epstein JI. Colorectal adenocarcinoma involving the prostate: report of 9 cases. Hum Pathol. 2007;38:1836ā€“41.

    PubMedĀ  Google ScholarĀ 

  50. Iczkowski KA, MacLennan GT, Bostwick DG. Atypical small acinar proliferation suspicious for malignancy in prostate needle biopsies: clinical significance in 33 cases. Am J Surg Pathol. 1997;21:1489ā€“95.

    PubMedĀ  CASĀ  Google ScholarĀ 

  51. Cheville JC, Reznicek MJ, Bostwick DG. The focus of ā€œatypical glands, suspicious for malignancyā€ in prostatic needle biopsy specimens: incidence, histologic features, and clinical follow-up of cases diagnosed in a community practice. Am J Clin Pathol. 1997;108:633ā€“40.

    PubMedĀ  CASĀ  Google ScholarĀ 

  52. Iczkowski KA, Bassler TJ, Schwob VS, et al. Diagnosis of ā€œsuspicious for malignancyā€ in prostate biopsies: predictive value for cancer. Urology. 1998;51:749ā€“58.

    PubMedĀ  CASĀ  Google ScholarĀ 

  53. Iczkowski KA, Chen HM, Yang XJ, et al. Prostate cancer diagnosed after initial biopsy with atypical small acinar proliferation suspicious for malignancy is similar to cancer found on initial biopsy. Urology. 2002;60:851ā€“4.

    PubMedĀ  Google ScholarĀ 

  54. Iczkowski KA, Bostwick DG. Atypical small acinar proliferation of the prostate: 16 yearsā€™ experience. Pathol Case Rev. 2014;19:147ā€“53.

    Google ScholarĀ 

  55. Pietzak EJ, Kabarriti AE, Mucksavage P, et al. The presence of high-grade prostatic intraepithelial neoplasia or atypia on prostate biopsy does not adversely affect prostatectomy outcomes for patients otherwise eligible for active surveillance. Urology. 2014;84:1442ā€“7.

    PubMedĀ  Google ScholarĀ 

  56. Van der Kwast TH, Evans A, Lockwood G, et al. Variability in diagnostic opinion among pathologists for single small atypical foci in prostate biopsies. Am J Surg Pathol. 2010;34:169ā€“77.

    PubMedĀ  Google ScholarĀ 

  57. Borboroglu PG, Sur RL, Roberts JL, et al. Repeat biopsy strategy in patients with atypical small acinar proliferation or high grade prostatic intraepithelial neoplasia on initial prostate needle biopsy. J Urol. 2001;166:866ā€“70.

    PubMedĀ  CASĀ  Google ScholarĀ 

  58. Moore CK, Karikehalli S, Nazeer T, et al. Prognostic significance of high grade prostatic intraepithelial neoplasia and atypical small acinar proliferation in the contemporary era. J Urol. 2005;173:70ā€“2.

    PubMedĀ  Google ScholarĀ 

  59. Amin MM, Jeyaganth S, Fahmy N, et al. Subsequent prostate cancer detection in patients with prostatic intraepithelial neoplasia or atypical small acinar proliferation. Can Urol Assoc J. 2007;1:245ā€“9.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  60. Lopez JI. Prostate adenocarcinoma detected after high-grade prostatic intraepithelial neoplasia or atypical small acinar proliferation. BJU Int. 2007;100:1272ā€“6.

    PubMedĀ  Google ScholarĀ 

  61. Montironi R, Mazzucchelli R. HER-2 expression and gene amplification in high-grade PIN and prostate cancer. Arch Ital Urol Androl. 2006;78:135ā€“9.

    PubMedĀ  Google ScholarĀ 

  62. Ploussard G, Plennevaux G, Allory Y, et al. High-grade prostatic intraepithelial neoplasia and atypical small acinar proliferation on initial 21-core extended biopsy scheme: incidence and implications for patient care and surveillance. World J Urol. 2009;27:587ā€“92.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  63. Gleason DF. Classification of prostatic carcinomas. Cancer Chemother Rep. 1966;50:125ā€“8.

    PubMedĀ  CASĀ  Google ScholarĀ 

  64. Moch H, Humphrey PA, Ulbright TM, Reuter VE. WHO classification of tumours of the urinary system and male genital organs. 4th ed. Lyon: International Agency for Research on Cancer; 2016.

    Google ScholarĀ 

  65. Epstein JI, Srigley J, Grignon D, Humphrey P, Association of Directors of Anatomic and Surgical Pathology. Recommendations for the reporting of prostate carcinoma. Hum Pathol. 2007;38:1305ā€“9.

    PubMedĀ  Google ScholarĀ 

  66. Epstein JI, Egevad L, Amin MB, et al. The 2014 International Society of Urological Pathology (ISUP) consensus conference on Gleason grading of prostatic carcinoma: definition of grading patterns and proposal for a new grading system. Am J Surg Pathol. 2016;40:244ā€“52.

    PubMedĀ  Google ScholarĀ 

  67. Liu JJ, Lichtensztajn DY, Gomez SL, et al. Nationwide prevalence of lymph node metastases in Gleason score 3 + 3 = 6 prostate cancer. Pathology. 2014;46:306ā€“10.

    PubMedĀ  Google ScholarĀ 

  68. Klein EA, Santiago-JimĆ©nez M, Yousefi K, et al. Molecular analysis of low grade prostate cancer using a genomic classifier of metastatic potential. J Urol. 2017;197:122ā€“8.

    PubMedĀ  CASĀ  Google ScholarĀ 

  69. Iczkowski KA, La Rosa FG. Gleason 6 cancer is still cancer. Oncology (Williston Park). 2014;28:22. 24ā€“29.

    Google ScholarĀ 

  70. Zhou M, Li J, Cheng L, et al. Diagnosis of ā€œpoorly formed glandsā€ Gleason pattern 4 prostatic adenocarcinoma on needle biopsy: an interobserver reproducibility study among urologic pathologists with recommendations. Am J Surg Pathol. 2015;39:1331ā€“9.

    PubMedĀ  Google ScholarĀ 

  71. Al-Hussain TO, Nagar MS, Epstein JI. Gleason pattern 5 is frequently underdiagnosed on prostate needle-core biopsy. Urology. 2012;79:178ā€“81.

    PubMedĀ  Google ScholarĀ 

  72. Fajardo DA, Miyamoto H, Miller JS, et al. Identification of Gleason pattern 5 on prostatic needle core biopsy: frequency of underdiagnosis and relation to morphology. Am J Surg Pathol. 2011;35:1706ā€“11.

    PubMedĀ  Google ScholarĀ 

  73. Shah RB, Li J, Cheng L. Diagnosis of Gleason pattern 5 prostate adenocarcinoma on core needle biopsy: an interobserver reproducibility study among urologic pathologists. Am J Surg Pathol. 2015;39:1242ā€“9.

    PubMedĀ  Google ScholarĀ 

  74. Kryvenko ON, Epstein JI. Prostate cancer grading: a decade after the 2005 modified Gleason system. Arch Pathol Lab Med. 2016;140:1140ā€“52.

    PubMedĀ  Google ScholarĀ 

  75. Epstein JI, Allsbrook WC, Amin MB, Egevad LL, The ISUP Grading Committee. The International Society of Urological Pathology (ISUP) consensus conference on Gleason grading of prostatic carcinoma. Am J Surg Pathol. 2005;29:1228ā€“42.

    PubMedĀ  Google ScholarĀ 

  76. Sauter G, Steurer S, Clauditz TS, et al. Clinical utility of quantitative Gleason grading in prostate biopsies and prostatectomy specimens. Eur Urol. 2016;69:592ā€“8.

    PubMedĀ  Google ScholarĀ 

  77. Iczkowski KA, Ferguson KL, Grier DD, et al. Adenoid cystic carcinoma of the prostate: clinicopathologic findings in 19 cases. Am J Surg Pathol. 2003;27:1523ā€“9.

    PubMedĀ  Google ScholarĀ 

  78. Berney DM. Low Gleason score prostatic adenocarcinomas are no longer viable entities. Histopathology. 2007;50:683ā€“90.

    PubMedĀ  CASĀ  Google ScholarĀ 

  79. Allsbrook WC Jr, Mangold KA, Johnson MH, et al. Interobserver reproducibility of Gleason grading of prostatic carcinoma: urologic pathologists. Hum Pathol. 2001;32:74ā€“80.

    PubMedĀ  Google ScholarĀ 

  80. Steinberg DM, Sauvageot J, Piantadosi S, et al. Correlation of prostate needle biopsy and radical prostatectomy Gleason grade in academic and community settings. Am J Surg Pathol. 1997;21:566ā€“76.

    PubMedĀ  CASĀ  Google ScholarĀ 

  81. Iczkowski KA, Bostwick DG. The pathologist as optimist: cancer grade deflation in prostatic needle biopsies (editorial). Am J Surg Pathol. 1998;22:1169ā€“70.

    PubMedĀ  CASĀ  Google ScholarĀ 

  82. Danneman D, Drevin L, Robinson D, et al. Gleason inflation 1998-2011: a registry study of 97,168 men. BJU Int. 2015;115:248ā€“55.

    PubMedĀ  CASĀ  Google ScholarĀ 

  83. Chen SD, Fava JL, Amin A. Gleason grading challenges in the diagnosis of prostate adenocarcinoma: experience of a single institution. Virchows Arch. 2016;468:213ā€“8.

    PubMedĀ  Google ScholarĀ 

  84. Gordetsky JB, Thomas JV, Nix JW, et al. Higher prostate cancer grade groups are detected in patients undergoing multiparametric MRI-targeted biopsy compared with standard biopsy. Am J Surg Pathol. 2017;41:101ā€“5.

    PubMedĀ  Google ScholarĀ 

  85. Sakr WA, Tefilli MV, Grignon DJ, et al. Gleason score 7 prostate cancer: a heterogeneous entity? Correlation with pathologic parameters and disease-free survival. Urology. 2000;56:730ā€“4.

    PubMedĀ  CASĀ  Google ScholarĀ 

  86. Perlis N, Sayyid R, Evans A, et al. Limitations in predicting organ-confined prostate cancer in patients with Gleason pattern 4 on biopsy: implications for active surveillance. J Urol. 2017;197:75ā€“83.

    PubMedĀ  Google ScholarĀ 

  87. Choy B, Pearce SM, Anderson BB, et al. Prognostic significance of percentage and architectural types of contemporary Gleason pattern 4 prostate cancer in radical prostatectomy. Am J Surg Pathol. 2016;40(10):1400ā€“6.

    PubMedĀ  Google ScholarĀ 

  88. Sadamin ET, Khani F, Diolombi M, et al. Interobserver reproducibility of percent Gleason 4 in prostatic adenocarcinoma on prostate biopsies. Am J Surg Pathol. 2016;40:1686ā€“92.

    Google ScholarĀ 

  89. Kweldam CF, Nieboer D, Algaba F, et al. Gleason grade 4 prostate adenocarcinoma patterns: an inter-observer agreement study among genitourinary pathologists. Histopathology. 2016;69:441ā€“9.

    PubMedĀ  Google ScholarĀ 

  90. Kryvenko ON, Epstein JI. Changes in prostate cancer grading: including a new patient-centric grading system. Prostate. 2016;76:427ā€“33.

    PubMedĀ  Google ScholarĀ 

  91. Pierorazio PM, Walsh PC, Partin AW, et al. Prognostic Gleason grade grouping: data based on the modified Gleason scoring system. BJU Int. 2013;111:753ā€“60.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  92. Epstein JI, Zelefsky MJ, Sjoberg DD, et al. A contemporary prostate cancer grading system: a validated alternative to the Gleason score. Eur Urol. 2016;69:428ā€“35.

    PubMedĀ  Google ScholarĀ 

  93. Egevad L, Delahunt B, Evans AJ, et al. International Society of Urologic Pathology (ISUP) grading of prostate cancer. Am J Surg Pathol. 2016;40:858ā€“61.

    PubMedĀ  Google ScholarĀ 

  94. Epstein JI. International Society of Urologic Pathology (ISUP) grading of prostate cancer: authorā€™s reply. Am J Surg Pathol. 2016;40:862ā€“4.

    PubMedĀ  Google ScholarĀ 

  95. O'Kelly F, Elamin S, Cahill A, et al. Characteristics of modern Gleason 9/10 prostate adenocarcinoma: a single tertiary centre experience within the Republic of Ireland. World J Urol. 2014;32:1067ā€“74.

    PubMedĀ  CASĀ  Google ScholarĀ 

  96. Tsao CK, Gray KP, Nakabayashi M, et al. Patients with biopsy Gleason 9 and 10 prostate cancer have significantly worse outcomes compared to patients with Gleason 8 disease. J Urol. 2015;194:91ā€“7.

    PubMedĀ  Google ScholarĀ 

  97. Harding-Jackson N, Kryvenko ON, Whittington EE, et al. Outcome of Gleason 3+5=8 prostate cancer diagnosed on needle biopsy: prognostic comparison with Gleason 4+4=8. J Urol. 2016;196:1076ā€“81.

    PubMedĀ  Google ScholarĀ 

  98. Flood TA, Schieda N, Keefe DT, et al. Utility of Gleason pattern 4 morphologies detected on transrectal ultrasound (TRUS)-guided biopsies for prediction of upgrading or upstaging in Gleason score 3 + 4 = 7 prostate cancer. Virchows Arch. 2016;469:313ā€“9.

    PubMedĀ  Google ScholarĀ 

  99. Kweldam CF, KĆ¼mmerlin IP, Nieboer D, et al. Prostate cancer outcomes of men with biopsy Gleason score 6 and 7 without cribriform or intraductal carcinoma. Eur J Cancer. 2016;66:26ā€“33.

    PubMedĀ  Google ScholarĀ 

  100. Tosoian JJ, Mamawala M, Epstein JI, et al. Intermediate and longer-term outcomes from a prospective active-surveillance program for favorable-risk prostate cancer. J Clin Oncol. 2015;33:3379ā€“85.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  101. Samaratunga H, Montironi R, True L, et al. International Society of Urological Pathology (ISUP) consensus conference on handling and staging of radical prostatectomy specimens. Working group 1: specimen handling. Mod Pathol. 2011;24:6ā€“15.

    PubMedĀ  Google ScholarĀ 

  102. Trpkov K, Gao Y, Hay R, et al. No residual cancer on radical prostatectomy after positive 10-core biopsy: incidence, biopsy findings, and DNA specimen identity analysis. Arch Pathol Lab Med. 2006;130:811ā€“6.

    PubMedĀ  Google ScholarĀ 

  103. Prayer-Galetti T, Gardiman M, Sacco E, et al. Finding of no tumor (pT0) in patients undergoing radical retropubic prostatectomy for clinically localized prostate cancer. Anal Quant Cytol Histol. 2007;29:79ā€“86.

    PubMedĀ  Google ScholarĀ 

  104. van Oort IM, Witjes JA, Kok DE, et al. The prognostic role of the pathological T2 subclassification for prostate cancer in the 2002 Tumor-Nodes-Metastasis staging system. BJU Int. 2008;102:438ā€“41.

    PubMedĀ  Google ScholarĀ 

  105. Kollerman J, Feek U, Muller H, et al. Nondetected tumor (pT0) after prolonged, neoadjuvant treatment of localized prostatic carcinoma. Eur Urol. 2000;38:714ā€“20.

    Google ScholarĀ 

  106. Descazeaud A, Zerbib M, Flam T, et al. Can pT0 stage of prostate cancer be predicted the before radical prostatectomy? Eur Urol. 2006;50:1248ā€“52.

    PubMedĀ  Google ScholarĀ 

  107. Montironi R, Cheng L, Lopez-Beltran A, et al. Stage pT0 in radical prostatectomy with no residual carcinoma and with a previous positive biopsy conveys a wrong message to clinicians and patients. Why is cancer not present in the radical prostatectomy specimen? Eur Urol. 2009;56:272ā€“4.

    PubMedĀ  Google ScholarĀ 

  108. Humphrey PA. Prostate tissue handling and sampling: transurethral prostatectomy (TURP). In: Humphrey PA, editor. Prostate pathology. Chicago: American Society of Clinical Pathology; 2005. p. 40ā€“4.

    Google ScholarĀ 

  109. Trpkov K, Thompson J, Kulaga A. How much tissue sampling is required when unsuspected minimal prostate carcinoma is identified on transurethral resection? Arch Pathol Lab Med. 2008;132:1313ā€“6.

    PubMedĀ  Google ScholarĀ 

  110. Chapter 58, Prostate. In: M. B. Amin et al. (eds.), AJCC cancer staging manual, 8th ed. Chicago: Springer, 2017. pp. 715ā€“726.

    Google ScholarĀ 

  111. Magi-Galluzzi C, Evans AJ, Delahunt B, et al. International Society of Urological Pathology (ISUP) consensus conference on handling and staging of radical prostatectomy specimens. Working group 3: extraprostatic extension, lymphovascular invasion and locally advanced disease. Mod Pathol. 2011;24:26ā€“38.

    PubMedĀ  Google ScholarĀ 

  112. van der Kwast TH, Amin MB, Billis A, et al. International Society of Urological Pathology (ISUP) consensus conference on handling and staging of radical prostatectomy specimens. Working group 2: T2 substaging and prostate cancer volume. Mod Pathol. 2011;24:16ā€“25.

    PubMedĀ  Google ScholarĀ 

  113. Ettel M, Kong M, Lee P, et al. Modification of the pT2 substage classification in prostate adenocarcinoma. Hum Pathol. 2016;56:57ā€“63.

    PubMedĀ  Google ScholarĀ 

  114. Nazeer T, Kee KH, Ro JY, et al. Intraprostatic adipose tissue: a study of 427 whole-mount radical prostatectomy specimens. Hum Pathol. 2009;40:538ā€“41.

    PubMedĀ  Google ScholarĀ 

  115. Berney DM, Wheeler TM, Grignon DJ, et al. International Society of Urological Pathology (ISUP) consensus conference on handling and staging of radical prostatectomy specimens. Working group 4: seminal vesicles and lymph nodes. Mod Pathol. 2011;24:39ā€“47.

    PubMedĀ  Google ScholarĀ 

  116. Ohori M, Scardino PT, Lapin SL, et al. The mechanisms and prognostic significance of seminal vesicle involvement by prostate cancer. Am J Surg Pathol. 1993;17:1252ā€“61.

    PubMedĀ  CASĀ  Google ScholarĀ 

  117. Luchini C, Fleischmann A, Boormans JL, et al. Extranodal extension of lymph node metastasis influences recurrence in prostate cancer: a systematic review and meta-analysis. Sci Rep. 2017; 24;7(1):2374.

    Google ScholarĀ 

  118. Ramos CG, Roehl KA, Antenor JA, et al. Percent carcinoma in prostatectomy specimen is associated with risk of recurrence after radical prostatectomy in patients with pathologically organ confined prostate cancer. J Urol. 2004;172:137ā€“40.

    PubMedĀ  Google ScholarĀ 

  119. Renshaw AA, Richie JP, Loughlin KR, et al. The greatest dimension of prostate carcinoma is a simple, inexpensive predictor of prostate specific antigen failure in radical prostatectomy specimens. Cancer. 1998;83:748ā€“52.

    PubMedĀ  CASĀ  Google ScholarĀ 

  120. Iczkowski KA, Lucia MS. Frequency of positive surgical margin at prostatectomy and its effect on patient outcome. Prostate Cancer. 2011;2011:673021. Epub 2011 Jun 9.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  121. Blute ML, Bostwick DG, Bergstralh EJ, et al. Anatomic site-specific positive margins in organ-confined prostate cancer and its impact on outcome after radical prostatectomy. Urology. 1997;50:733ā€“9.

    PubMedĀ  CASĀ  Google ScholarĀ 

  122. Wheeler TM, Dillioglugil O, Kattan MW, et al. Clinical and pathological significance of the level and extent of capsular invasion in clinical stage T1-2 prostate cancer. Hum Pathol. 1998;29:856ā€“62.

    PubMedĀ  CASĀ  Google ScholarĀ 

  123. Tan PH, Cheng L, Srigley JR, et al. International Society of Urological Pathology (ISUP) consensus conference on handling and staging of radical prostatectomy specimens. Working group 5: surgical margins. Mod Pathol. 2011;24:48ā€“57.

    PubMedĀ  Google ScholarĀ 

  124. Whalen MJ, Shapiro EY, Rothberg MB, et al. Close surgical margins after radical prostatectomy mimic biochemical recurrence rates of positive margins. Urol Oncol. 2015;33(11):494.e9ā€“494.e14.

    Google ScholarĀ 

  125. Paluru S, Epstein JI. Does the distance between tumor and margin in radical prostatectomy specimens correlate with prognosis: relation to tumor location. Hum Pathol. 2016;30:11ā€“15.

    Google ScholarĀ 

  126. Koppie TM, Bianco FJ, Kuroiwa K, et al. The clinical features of anterior prostate cancers. BJU Int. 2006;98:1167ā€“71.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  127. Kryvenko ON, Lyapichev K, Chinea FM, et al. Radical prostatectomy findings in white Hispanic/Latino men with NCCN very low-risk prostate cancer detected by template biopsy. Am J Surg Pathol. 2016;40:1125ā€“32.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  128. Kanthabalan A, Emberton M, Ahmed HU. Biopsy strategies for selecting patients for focal therapy for prostate cancer. Curr Opin Urol. 2014;24:209ā€“17.

    PubMedĀ  Google ScholarĀ 

  129. Iczkowski KA, Hossain D, Torkko KC, et al. Preoperative prediction of unifocal, unilateral, margin-negative, and small volume prostate cancer. Urology. 2008;71:1166ā€“71.

    PubMedĀ  Google ScholarĀ 

  130. Hawken SR, Womble PR, Herrel LA, et al. Understanding the performance of active surveillance selection criteria in diverse urology practices. J Urol. 2015;194:1253ā€“7.

    PubMedĀ  Google ScholarĀ 

  131. Egevad L, Engstrom K, Wester K, et al. Heterogeneity of DNA ploidy in prostate cancer. J Urol Pathol. 1999;10:23ā€“37.

    Google ScholarĀ 

  132. Iczkowski KA, Torkko KC, Kotnis GR, et al. Digital quantification of five high-grade prostate cancer patterns, including the cribriform pattern, and their association with adverse outcome. Am J Clin Pathol. 2011;136:98.

    PubMedĀ  Google ScholarĀ 

  133. Kryvenko ON, Gupta NS, Virani N, et al. Gleason score 7 adenocarcinoma of the prostate with lymph node metastases: analysis of 184 radical prostatectomy specimens. Arch Pathol Lab Med. 2013;137:61.

    Google ScholarĀ 

  134. Dong F, Yang P, Wang C, et al. Architectural heterogeneity and cribriform pattern predict adverse clinical outcome for Gleason grade 4 prostatic adenocarcinoma. Am J Surg Pathol. 2013;37:1855ā€“61.

    PubMedĀ  Google ScholarĀ 

  135. Trudel D, Downes MR, Sykes J, et al. Prognostic impact of intraductal carcinoma and large cribriform carcinoma architecture after prostatectomy in a contemporary cohort. Eur J Cancer. 2014;50:1610ā€“6.

    PubMedĀ  Google ScholarĀ 

  136. Sarbay BC, Kir G, Topal CS, et al. Significance of the cribriform pattern in prostatic adenocarcinomas. Pathol Res Pract. 2014;210:554ā€“7.

    PubMedĀ  Google ScholarĀ 

  137. Kir G, Sarbay BC, GĆ¼mĆ¼ÅŸ E, et al. The association of the cribriform pattern with outcome for prostatic adenocarcinomas. Pathol Res Pract. 2014;210:640ā€“4.

    PubMedĀ  CASĀ  Google ScholarĀ 

  138. Siadat F, Sykes J, Zlotta AR, et al. Not all Gleason pattern 4 prostate cancers are created equal: a study of latent prostatic carcinomas in a cystoprostatectomy and autopsy series. Prostate. 2015;75:1277ā€“84.

    PubMedĀ  Google ScholarĀ 

  139. Keefe DT, Schieda N, El Hallani S, et al. Cribriform morphology predicts upstaging after radical prostatectomy in patients with Gleason score 3 + 4 = 7 prostate cancer at transrectal ultrasound (TRUS)-guided needle biopsy. Virchows Arch. 2015;467:437ā€“42.

    PubMedĀ  Google ScholarĀ 

  140. Kweldam CF, Wildhagen MF, Steyerberg EW, et al. Cribriform growth is highly predictive for postoperative metastasis and disease-specific death in Gleason score 7 prostate cancer. Mod Pathol. 2015;28:457ā€“64.

    PubMedĀ  Google ScholarĀ 

  141. Kweldam CF, KĆ¼mmerlin IP, Nieboer D, et al. Disease-specific survival of patients with invasive cribriform and intraductal prostate cancer at diagnostic biopsy. Mod Pathol. 2016; https://doi.org/10.1038/modpathol.2016.49. (Epub ahead of print).

  142. McKenney JK, Wei W, Hawley S, et al. Histologic grading of prostatic adenocarcinoma can be further optimized: analysis of the relative prognostic strength of individual architectural patterns in 1275 patients from the Canary Retrospective Cohort. Am J Surg Pathol. 2016;40:1439ā€“56.

    PubMedĀ  Google ScholarĀ 

  143. Rhamy RK, Buchanan RD, Spalding MJ. Intraductal carcinoma of the prostate gland. Trans Am Assoc Genitourin Surg. 1972;64:61ā€“8.

    PubMedĀ  CASĀ  Google ScholarĀ 

  144. Iczkowski KA. Intraductal carcinoma of the prostate: emerging support for a unique diagnostic entity. Pathol Case Rev. 2014;19:178ā€“83.

    Google ScholarĀ 

  145. Haffner MC, Weier C, Xu MM, et al. Molecular evidence that invasive adenocarcinoma can mimic prostatic intraepithelial neoplasia (PIN) and intraductal carcinoma through retrograde glandular colonization. J Pathol. 2016;238:31ā€“41.

    PubMedĀ  CASĀ  Google ScholarĀ 

  146. Lotan TL, Gumuskaya B, Rahimi H, et al. Cytoplasmic PTEN protein loss distinguishes intraductal carcinoma of the prostate from high-grade prostatic intraepithelial neoplasia. Mod Pathol. 2013;26:587ā€“603.

    PubMedĀ  CASĀ  Google ScholarĀ 

  147. Shah RB, Magi-Galluzzi C, Han B, et al. Atypical cribriform lesions of the prostate: relationship to prostatic carcinoma and implication for diagnosis in prostate biopsies. Am J Surg Pathol. 2010;34:470ā€“7.

    PubMedĀ  Google ScholarĀ 

  148. Iczkowski KA, et al. Intraductal carcinoma of the prostate: interobserver reproducibility survey of 39 urologic pathologists. Ann Diagn Pathol. 2014;18:333ā€“42.

    PubMedĀ  Google ScholarĀ 

  149. Miyai K, Kristiansen A, Egevad L, et al. Seminal vesicle intraepithelial involvement by prostate cancer: putative mechanism and clinicopathological significance. Hum Pathol. 2014;45:1805ā€“12.

    PubMedĀ  Google ScholarĀ 

  150. Bostwick DG, Kindrachuk RW, Rouse RV. Prostatic adenocarcinoma with endometrioid features. Clinical, pathologic, and ultrastructural findings. Am J Surg Pathol. 1985;9:595ā€“609.

    PubMedĀ  CASĀ  Google ScholarĀ 

  151. Seipel AH, Delahunt B, Samaratunga H, et al. Diagnostic criteria for ductal adenocarcinoma of the prostate: interobserver variability among 20 expert uropathologists. Histopathology. 2014;65:216ā€“27.

    PubMedĀ  Google ScholarĀ 

  152. Bock BJ, Bostwick DG. Does prostatic ductal adenocarcinoma exist? Am J Surg Pathol. 1999;23:781ā€“5.

    PubMedĀ  CASĀ  Google ScholarĀ 

  153. Ali TZ, Epstein JI. Basal cell carcinoma of the prostate: a clinicopathologic study of 29 cases. Am J Surg Pathol. 2007;31:697ā€“705.

    PubMedĀ  Google ScholarĀ 

  154. Iczkowski KA, Montironi R. Adenoid cystic/basal cell carcinoma of the prostate strongly expresses HER-2/neu. J Clin Pathol. 2006;59:1327ā€“30.

    PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  155. Zhang M, Pettaway C, Vikram R, Tamboli P. Adenoid cystic carcinoma of the urethra/Cowperā€™s gland with concurrent high-grade prostatic adenocarcinoma: a detailed clinico-pathologic case report and review of the literature. Hum Pathol. 2016;58:138ā€“44.

    PubMedĀ  CASĀ  Google ScholarĀ 

  156. di Sant'Agnese PA, de Mesy Jensen KL. Neuroendocrine differentiation in prostatic carcinoma. Hum Pathol. 1987;18:849ā€“56.

    PubMedĀ  Google ScholarĀ 

  157. Bostwick DG, Dousa MK, Crawford BG, Wollan PC. Neuroendocrine differentiation in prostatic intraepithelial neoplasia and adenocarcinoma. Am J Surg Pathol. 1994;18:1240ā€“6.

    PubMedĀ  CASĀ  Google ScholarĀ 

  158. Epstein JI, Amin MB, Beltran H, et al. Proposed morphologic classification of prostate cancer with neuroendocrine differentiation. Am J Surg Pathol. 2014;38:756ā€“67.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  159. Deorah S, Rao MB, Raman R, et al. Survival of patients with small cell carcinoma of the prostate during 1973-2003: a population-based study. BJU Int. 2012;109:824ā€“30.

    PubMedĀ  Google ScholarĀ 

  160. Evans AJ, Humphrey PA, Belani J, et al. Large cell neuroendocrine carcinoma of prostate: a clinicopathologic summary of 7 cases of a rare manifestation of advanced prostate cancer. Am J Surg Pathol. 2006;30:684ā€“93.

    PubMedĀ  Google ScholarĀ 

  161. Bassler TJ, Orozco R, Bassler IC, et al. Adenosquamous carcinoma of the prostate: case report with DNA analysis, immunohistochemistry, and literature review. Urology. 1999;53:832ā€“4.

    PubMedĀ  Google ScholarĀ 

  162. Munoz F, Franco P, Ciammella P, et al. Squamous cell carcinoma of the prostate: long-term survival after combined chemo-radiation. Radiat Oncol. 2007;2:15.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  163. Marcus DM, Goodman M, Jani AB, et al. A comprehensive review of incidence and survival in patients with rare histological variants of prostate cancer in the United States from 1973 to 2008. Prostate Cancer Prostatic Dis. 2012;15:283ā€“8.

    PubMedĀ  CASĀ  Google ScholarĀ 

  164. Abbott D, Iczkowski KA. Squamous cell carcinoma of the prostate with concomitant adenocarcinoma in the absence of prior androgen deprivation therapy. Case Rev Clin Pathol. 2016;3:60ā€“3.

    Google ScholarĀ 

  165. Crook JM, Perry GA, Robertson S, Esche BA. Routine prostate biopsies following radiotherapy for prostate cancer: results for 226 patients. Urology. 1995;45:624ā€“31.

    PubMedĀ  CASĀ  Google ScholarĀ 

  166. Siders DB, Lee F. Histologic changes of irradiated prostatic carcinoma diagnosed by transrectal ultrasound. Hum Pathol. 1992;23:344ā€“51.

    PubMedĀ  CASĀ  Google ScholarĀ 

  167. Iczkowski KA. Effect of radiotherapy on non-neoplastic and malignant prostate. Open Pathol J. 2009;3:64ā€“73.

    CASĀ  Google ScholarĀ 

  168. Helpap B, Bonkhoff H, Cockett A, et al. Relationship between atypical adenomatous hyperplasia (AAH), prostatic intraepithelial neoplasia (PIN) and prostatic adenocarcinoma. Pathologica. 1997;89:288ā€“300.

    PubMedĀ  CASĀ  Google ScholarĀ 

  169. Iczkowski KA, Qiu J, Qian J, et al. The dual 5Ī±-reductase inhibitor dutasteride induces atrophic changes and decreases relative cancer volume in human prostate. Urology. 2005;65:76ā€“82.

    PubMedĀ  Google ScholarĀ 

  170. Gleave M, Qian J, Andreou C, et al. The effects of the dual 5alpha-reductase inhibitor dutasteride on localized prostate cancer--results from a 4-month pre-radical prostatectomy study. Prostate. 2006;66:1674ā€“85.

    PubMedĀ  CASĀ  Google ScholarĀ 

  171. Ellison E, Ferguson J, Zincke H, et al. ā€œNucleolus-poorā€ clear cell adenocarcinoma of the prostate: a distinctive histologic variant in patients receiving total androgen blockade [abstract]. Mod Pathol. 1993;59A

    Google ScholarĀ 

  172. Ferguson J, Zincke H, Ellison E, et al. Decrease of prostatic intraepithelial neoplasia following androgen deprivation therapy in patients with stage T3 carcinoma treated by radical prostatectomy. Urology. 1994;44:91ā€“5.

    PubMedĀ  CASĀ  Google ScholarĀ 

  173. Gaudin PB, Rosai J, Epstein JI. Sarcomas and related proliferative lesions of specialized prostatic stroma: a clinicopathologic study of 22 cases. Am J Surg Pathol. 1998;22:148ā€“62.

    PubMedĀ  CASĀ  Google ScholarĀ 

  174. Bostwick DG, Hossain D, Qian J, et al. Phyllodes tumor of the prostate: long-term follow up study of 23 cases. J Urol. 2004;172:894ā€“9.

    PubMedĀ  Google ScholarĀ 

  175. Herawi M, Epstein JI. Specialized stromal tumors of the prostate: a clinicopathologic study of 50 cases. Am J Surg Pathol. 2006;30:694ā€“704.

    PubMedĀ  Google ScholarĀ 

  176. Mobbs BG, Liu Y. Immunohistochemical localization of progesterone receptor in benign and malignant human prostate. Prostate. 1990;16:245ā€“51.

    PubMedĀ  CASĀ  Google ScholarĀ 

  177. Hossain D, Meiers I, Qian J, et al. Prostatic stromal hyperplasia with atypia: follow-up study of 18 cases. Arch Pathol Lab Med. 2008;132:1729ā€“33.

    PubMedĀ  Google ScholarĀ 

  178. Wang J, Wang F. The impact of radical prostatectomy on the survival of patients with carcinosarcoma of the prostate. J Cancer Ther. 2011;2:475ā€“80.

    CASĀ  Google ScholarĀ 

  179. Sountoulides P, Koletsas N, Kikidakis D, et al. Secondary malignancies following radiotherapy for prostate cancer. Ther Adv Urol. 2010;2:119ā€“25.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  180. Siegele BJ, Iczkowski KA, La Rosa FG. Carcinosarcoma of the prostate in a patient with previous prostatic adenocarcinoma, status post brachytherapy. Int J Clin Exp Pathol. 2016;9(7):XXXā€“XXX, in press.

    Google ScholarĀ 

  181. Miedler JD, MacLennan GT. Leiomyosarcoma of the prostate. J Urol. 2007;178:668.

    PubMedĀ  Google ScholarĀ 

  182. Asahina M, Saito T, Arakawa A, et al. A case of primary spindle cell variant of embryonal rhabdomyosarcoma of the prostate. Int J Clin Exp Pathol. 2014;7(8):5181ā€“5.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  183. Schildhaus HU, Lokka S, Fenner W, et al. Spindle cell embryonal rhabdomyosarcoma of the prostate in an adult patient - case report and review of clinicopathological features. Diagn Pathol. 2016;11:56.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  184. Anagnostou E, Miliaras D, Panagiotakopoulos V. Diagnosis of gastrointestinal stromal tumor (GIST) on transurethral resection of the prostate: a case report and review of the literature. Int J Surg Pathol. 2011;19:632ā€“6.

    PubMedĀ  Google ScholarĀ 

  185. Bostwick DG, Iczkowski KA, Amin MB, et al. Malignant lymphoma involving the prostate: report of 62 cases. Cancer. 1998;83:732ā€“8.

    PubMedĀ  CASĀ  Google ScholarĀ 

  186. Chu PG, Huang Q, Weiss LM. Incidental and concurrent malignant lymphomas discovered at the time of prostatectomy and prostate biopsy: a study of 29 cases. Am J Surg Pathol. 2005;29:693ā€“9.

    PubMedĀ  Google ScholarĀ 

  187. Quien ET, Wallach B, Sandhaus L, et al. Primary extramedullary leukemia of the prostate: case report and review of the literature. Am J Hematol. 1996;53:267ā€“71.

    PubMedĀ  CASĀ  Google ScholarĀ 

  188. Spethmann S, Heuer R, Hopfer H, et al. Myeloid sarcoma of the prostate as first clinical manifestation of acute myeloid leukaemia. Lancet Oncol. 2004;5:62ā€“3.

    PubMedĀ  Google ScholarĀ 

  189. Anderson GA, Braaten K. Prostatic extramedullary leukemia as a first site of relapse of acute nonlymphocytic leukemia. Urol Oncol. 2005;23:419ā€“21.

    PubMedĀ  Google ScholarĀ 

  190. Andrews C, Humphrey PA. Utility of ERG versus AMACR expression in diagnosis of minimal adenocarcinoma of the prostate in needle biopsy tissue. Am J Surg Pathol. 2014;38:1007ā€“12.

    PubMedĀ  Google ScholarĀ 

  191. Lee WH, Morton RA, Epstein JI, et al. Cytidine methylation of regulatory sequences near the pi-class glutathione S-transferase gene accompanies human prostatic carcinogenesis. Proc Natl Acad Sci U S A. 1994;91:11733ā€“7.

    PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  192. van Neste L, Herman JG, Otto G, et al. The epigenetic promise for prostate cancer diagnosis. Prostate. 2012;72:1248ā€“61.

    PubMedĀ  Google ScholarĀ 

  193. Stewart GD, Leander VN, Delvenne P, et al. Clinical utility of an epigenetic assay to detect occult prostate cancer in histopathologically negative biopsies: results of the MATLOC study. J Urol. 2013;189:1110ā€“6.

    PubMedĀ  Google ScholarĀ 

  194. Kron KJ, Liu L, Pethe VV, et al. DNA methylation of HOXD3 as a marker of prostate cancer progression. Lab Investig. 2010;90:1060ā€“7.

    PubMedĀ  CASĀ  Google ScholarĀ 

  195. Crawford ED, Rove KO, Trabulsi EJ, et al. Diagnostic performance of PCA3 to detect prostate cancer in men with increased prostate specific antigen: a prospective study of 1,962 cases. J Urol. 2012;188:1726ā€“31.

    PubMedĀ  Google ScholarĀ 

  196. Whitman EJ, Groskopf J, Ali A, Chen Y, Blase A, Furusato B, Petrovics G, Ibrahim M, Elsamanoudi S, Cullen J, Sesterhenn IA, Brassell S, Rittenhouse H, Srivastava S, McLeod DG. PCA3 score before radical prostatectomy predicts extracapsular extension and tumor volume. J Urol. 2008;180:1975ā€“8.

    PubMedĀ  Google ScholarĀ 

  197. http://mctp.med.umich.edu/physicians/molecular-testing-lab/available-tests (Accessed 9-21-14).

  198. Young A, Palanisamy N, Siddiqui J, Wood DP, et al. Correlation of urine TMPRSS2:ERG and PCA3 to ERG+ and total prostate cancer burden. Am J Clin Pathol. 2012;138:685ā€“96.

    PubMedĀ  Google ScholarĀ 

  199. Prostate health index: http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3943368/. Accessed 5 July 2014.

  200. Fossati N, Lazzeri M, Haese A, et al. Clinical performance of serum isoform [āˆ’2]proPSA (p2PSA) and its derivatives, %p2PSA and the prostate health index, in men <60 years: results from a multicentric European prospective study. BJU Int. 2015;115:913ā€“20.

    PubMedĀ  CASĀ  Google ScholarĀ 

  201. Rhodes T, Jacobson DJ, McGree ME, et al. Longitudinal changes of benign prostate-specific antigen and (āˆ’2)proprostate-specific antigen in seven years in a community-based sample of men. Urology. 2012;79:655ā€“61.

    PubMedĀ  Google ScholarĀ 

  202. Maccini M, Westfall N, van Bokhoven A, et al. The 4KScore can potentially reduce unnecessary prostate biopsies in men with suspicious PSA in a screening population. Poster #M65 presented at the 95th Annual Meeting, South Central Section of the American Urologic Association, Colorado Springs, Oct 1, 2016.

    Google ScholarĀ 

  203. Khan AP, Rajendiran TM, Ateeq B, et al. The role of sarcosine metabolism in prostate cancer progression. Neoplasia. 2013;15:491ā€“501.

    PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  204. http://www.bostwicklaboratories.com/Company/News/Metabolon-and-Bostwick-Laboratories-Announce-Marke.aspx. Accessed 8-12-14.

  205. Carver BS, Tran J, Gopalan A, et al. Aberrant ERG expression cooperates with loss of PTEN to promote cancer progression in the prostate. Nat Genet. 2009;41:619ā€“24.

    PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  206. Tomlins SA, Mehra R, Rhodes DR, et al. TMPRSS2:ETV4 gene fusions define a third molecular subtype of prostate cancer. Cancer Res. 2006;66:3396ā€“400.

    PubMedĀ  CASĀ  Google ScholarĀ 

  207. Klezovitch O, Risk M, Coleman I, et al. A causal role for ERG in neoplastic transformation of prostate epithelium. Proc Natl Acad Sci U S A. 2008;105:2105ā€“10.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  208. Park K, Tomlins SA, Mudaliar KM, et al. Antibody-based detection of ERG rearrangement-positive prostate cancer. Neoplasia. 2010;12:590ā€“8.

    PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  209. Yoshimoto M, Joshua AM, Cunha IW, et al. Absence of TMPRSS2:ERG fusions and PTEN losses in prostate cancer is associated with a favorable outcome. Mod Pathol. 2008;21:1451ā€“60.

    PubMedĀ  CASĀ  Google ScholarĀ 

  210. Hoogland AM, Jenster G, van Weerden WM, et al. ERG immunohistochemistry is not predictive for PSA recurrence, local recurrence or overall survival after radical prostatectomy for prostate cancer. Mod Pathol. 2012;25:471ā€“9.

    PubMedĀ  CASĀ  Google ScholarĀ 

  211. McCall P, Witton CJ, Grimsley S, et al. Is PTEN loss associated with clinical outcome measures in human prostate cancer? Br J Cancer. 2008;99:1296ā€“301.

    PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  212. Krohn A, Diedler T, Burkhardt L, et al. Genomic deletion of PTEN is associated with tumor progression and early PSA recurrence in ERG fusion-positive and fusion-negative prostate cancer. Am J Pathol. 2012;181:401ā€“12.

    PubMedĀ  CASĀ  Google ScholarĀ 

  213. Mithal P, Allott E, Gerber L, et al. PTEN loss in biopsy tissue predicts poor clinical outcomes in prostate cancer. Int J Urol. 2014;21:1209ā€“14.

    PubMedĀ  CASĀ  Google ScholarĀ 

  214. Lotan TL, Gurel B, Sutcliffe S, et al. PTEN protein loss by immunostaining: analytic validation and prognostic indicator for a high risk surgical cohort of prostate cancer patients. Clin Cancer Res. 2011;17:6563ā€“73.

    PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  215. Verhagen PC, van Duijn PW, Hermans KG, et al. The PTEN gene in locally progressive prostate cancer is preferentially inactivated by bi-allelic gene deletion. J Pathol. 2006;208:699ā€“707.

    PubMedĀ  CASĀ  Google ScholarĀ 

  216. Han B, Mehra R, Lonigro RJ, et al. Fluorescence in situ hybridization study shows association of PTEN deletion with ERG rearrangement during prostate cancer progression. Mod Pathol. 2009;22:1083ā€“93.

    PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  217. Lahdensuo K, Erickson A, Saarinen I, et al. Loss of PTEN expression in ERG-negative prostate cancer predicts secondary therapies and leads to shorter disease-specific survival time after radical prostatectomy. Mod Pathol. 2016;29:1565ā€“74.

    PubMedĀ  CASĀ  Google ScholarĀ 

  218. Fine SW, Gopalan A, Leversha MA, et al. TMPRSS2-ERG gene fusion is associated with low Gleason scores and not with high-grade morphological features. Mod Pathol. 2010;23:1325ā€“33.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  219. Dawkins HJ, Sellner LN, Turbett GR, et al. Distinction between intraductal carcinoma of the prostate (IDC-P), high-grade dysplasia (PIN), and invasive prostatic adenocarcinoma, using molecular markers of cancer progression. Prostate. 2000;44:265ā€“70.

    PubMedĀ  CASĀ  Google ScholarĀ 

  220. Schneider TM, Osunkoya AO. ERG expression in intraductal carcinoma of the prostate: comparison with adjacent conventional acinar prostatic adenocarcinoma. Mod Pathol. 2013;25:247A.

    Google ScholarĀ 

  221. Reid AH, Attard G, Ambroisine L, et al. Molecular characterisation of ERG, ETV1 and PTEN gene loci identifies patients at low and high risk of death from prostate cancer. Br J Cancer. 2010;102:678ā€“84.

    PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  222. Yoshimoto M, Ding K, Sweet JM, et al. PTEN losses exhibit heterogeneity in multifocal prostatic adenocarcinoma and are associated with higher Gleason score. Mod Pathol. 2013;26:435ā€“47.

    PubMedĀ  CASĀ  Google ScholarĀ 

  223. Bhalla R, Kunju LP, Tomlins SA, et al. Novel dual-color immunohistochemical methods for detecting ERG-PTEN and ERG-SPINK1 status in prostate carcinoma. Mod Pathol. 2013;26:835ā€“48.

    PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  224. Chaux A, Peskoe SB, Gonzalez-Roibon N, et al. Loss of PTEN expression is associated with increased risk of recurrence after prostatectomy for clinically localized prostate cancer. Mod Pathol. 2012;25:1543ā€“9.

    PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  225. Lotan TL, Gurel B, Sutcliffe S, et al. PTEN protein loss: analytic validation and prognostic indicator for a high risk surgical cohort of prostate cancer patients. Clin Cancer Res. 2011;17:6563ā€“73.

    PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  226. Genomic Health Announces Data Reinforcing Robustness of Oncotype DXĀ® Prostate Cancer Test at 2014 American Urological Association (AUA) Annual Meeting http://investor.genomichealth.com/releaseDetail.cfm?releaseID=849215. Accessed 9-21-14.

  227. Subik MK, Gordetsky J, Yao JL, di Santā€™Agnese PA, Miyamoto H. Frozen section assessment in testicular and paratesticular lesions suspicious for malignancy: its role in preventing unnecessary orchiectomy. Hum Pathol. 2012;43(9):1514ā€“9.

    PubMedĀ  Google ScholarĀ 

  228. Leroy X, Rigot JM, Aubert S, Ballereau C, Gosselin B. Value of frozen section examination for the management of nonpalpable incidental testicular tumors. Eur Urol. 2003;44(4):458ā€“60.

    PubMedĀ  Google ScholarĀ 

  229. Connolly SS, Dā€™Arcy FT, Bredin HC, Callaghan J, Corcoran MO. Value of frozen section analysis with suspected testicular malignancy. Urology. 2006;67(1):162ā€“5.

    PubMedĀ  Google ScholarĀ 

  230. Amin MB, Greene FL, Edge S, Byrd DB, Brookland RK. AJCC cancer staging manual. New York: Springer; 2017. 1024 p.

    Google ScholarĀ 

  231. Aparicio J, Maroto P, Garcia del Muro X, Sanchez-Munoz A, Guma J, Margeli M, et al. Prognostic factors for relapse in stage I seminoma: a new nomogram derived from three consecutive, risk-adapted studies from the Spanish Germ Cell Cancer Group (SGCCG). Ann Oncol. 2014;25(11):2173ā€“8.

    PubMedĀ  CASĀ  Google ScholarĀ 

  232. Chung P, Daugaard G, Tyldesley S, Atenafu EG, Panzarella T, Kollmannsberger C, et al. Evaluation of a prognostic model for risk of relapse in stage I seminoma surveillance. Cancer Med. 2015;4(1):155ā€“60.

    PubMedĀ  Google ScholarĀ 

  233. Warde P, Specht L, Horwich A, Oliver T, Panzarella T, Gospodarowicz M, et al. Prognostic factors for relapse in stage I seminoma managed by surveillance: a pooled analysis. J Clin Oncol. 2002;20(22):4448ā€“52.

    PubMedĀ  Google ScholarĀ 

  234. Bettocchi C, Coker CB, Deacon J, Parkinson C, Pryor JP. A review of testicular intratubular germ cell neoplasia in infertile men. J Androl. 1994;15(Suppl):14Sā€“6S.

    PubMedĀ  Google ScholarĀ 

  235. von der Maase H, Rorth M, Walbom-Jorgensen S, Sorensen BL, Christophersen IS, Hald T, et al. Carcinoma in situ of contralateral testis in patients with testicular germ cell cancer: study of 27 cases in 500 patients. Br Med J. 1986;293(6559):1398ā€“401.

    Google ScholarĀ 

  236. Giwercman A, Bruun E, Frimodt-Moller C, Skakkebaek NE. Prevalence of carcinoma in situ and other histopathological abnormalities in testes of men with a history of cryptorchidism. J Urol. 1989;142(4):998ā€“1001. discussion 01-2.

    PubMedĀ  CASĀ  Google ScholarĀ 

  237. Hornak M, Pauer M, Bardos A Jr, Ondrus D. The incidence of carcinoma in situ in postpubertal undescended testis. Int Urol Nephrol. 1987;19(3):321ā€“5.

    PubMedĀ  CASĀ  Google ScholarĀ 

  238. Rogers E, Teahan S, Gallagher H, Butler MR, Grainger R, McDermott TE, et al. The role of orchiectomy in the management of postpubertal cryptorchidism. J Urol. 1998;159(3):851ā€“4.

    PubMedĀ  CASĀ  Google ScholarĀ 

  239. Muller J, Skakkebaek NE. Testicular carcinoma in situ in children with the androgen insensitivity (testicular feminisation) syndrome. Br Med J. 1984;288(6428):1419ā€“20.

    CASĀ  Google ScholarĀ 

  240. Williams JC, Merguerian PA, Schned AR, Amdur RJ. Bilateral testicular carcinoma in situ in persistent mullerian duct syndrome: a case report and literature review. Urology. 1994;44(4):595ā€“8.

    PubMedĀ  CASĀ  Google ScholarĀ 

  241. Osterlind A, Berthelsen JG, Abildgaard N, Hansen SO, Hjalgrim H, Johansen B, et al. Risk of bilateral testicular germ cell cancer in Denmark: 1960-1984. J Natl Cancer Inst. 1991;83(19):1391ā€“5.

    PubMedĀ  CASĀ  Google ScholarĀ 

  242. Wanderas EH, Fossa SD, Tretli S. Risk of a second germ cell cancer after treatment of a primary germ cell cancer in 2201 Norwegian male patients. Eur J Cancer. 1997;33(2):244ā€“52.

    PubMedĀ  CASĀ  Google ScholarĀ 

  243. Skakkebaek NE. Carcinoma in situ of the testis: frequency and relationship to invasive germ cell tumours in infertile men. Histopathology. 2002;41(3A):5ā€“18.

    PubMedĀ  CASĀ  Google ScholarĀ 

  244. Nistal M, Codesal J, Paniagua R. Carcinoma in situ of the testis in infertile men. A histological, immunocytochemical, and cytophotometric study of DNA content. J Pathol. 1989;159(3):205ā€“10.

    PubMedĀ  CASĀ  Google ScholarĀ 

  245. Daugaard G, Rorth M, von der Maase H, Skakkebaek NE. Management of extragonadal germ-cell tumors and the significance of bilateral testicular biopsies. Ann Oncol. 1992;3(4):283ā€“9.

    PubMedĀ  CASĀ  Google ScholarĀ 

  246. Bohle A, Studer UE, Sonntag RW, Scheidegger JR. Primary or secondary extragonadal germ cell tumors? J Urol. 1986;135(5):939ā€“43.

    PubMedĀ  CASĀ  Google ScholarĀ 

  247. Elzinga-Tinke JE, Sirre ME, Looijenga LH, van Casteren N, Wildhagen MF, Dohle GR. The predictive value of testicular ultrasound abnormalities for carcinoma in situ of the testis in men at risk for testicular cancer. Int J Androl. 2010;33(4):597ā€“603.

    PubMedĀ  CASĀ  Google ScholarĀ 

  248. Rud CN, Daugaard G, Rajpert-De Meyts E, Skakkebaek NE, Petersen JH, Jorgensen N. Sperm concentration, testicular volume and age predict risk of carcinoma in situ in contralateral testis of men with testicular germ cell cancer. J Urol. 2013;190(6):2074ā€“80.

    PubMedĀ  Google ScholarĀ 

  249. Dieckmann KP, Kulejewski M, Pichlmeier U, Loy V. Diagnosis of contralateral testicular intraepithelial neoplasia (TIN) in patients with testicular germ cell cancer: systematic two-site biopsies are more sensitive than a single random biopsy. Eur Urol. 2007;51(1):175ā€“83. discussion 83-5.

    PubMedĀ  Google ScholarĀ 

  250. van Casteren NJ, Boellaard WP, Dohle GR, Weber RF, Kuizinga MC, Stoop H, et al. Heterogeneous distribution of ITGCNU in an adult testis: consequences for biopsy-based diagnosis. Int J Surg Pathol. 2008;16(1):21ā€“4.

    PubMedĀ  Google ScholarĀ 

  251. Hoei-Hansen CE, Holm M, Rajpert-De Meyts E, Skakkebaek NE. Histological evidence of testicular dysgenesis in contralateral biopsies from 218 patients with testicular germ cell cancer. J Pathol. 2003;200(3):370ā€“4.

    PubMedĀ  Google ScholarĀ 

  252. Berney DM, Lee A, Shamash J, Oliver RT. The association between intratubular seminoma and invasive germ cell tumors. Hum Pathol. 2006;37(4):458ā€“61.

    PubMedĀ  CASĀ  Google ScholarĀ 

  253. Berney DM, Lee A, Randle SJ, Jordan S, Shamash J, Oliver RT. The frequency of intratubular embryonal carcinoma: implications for the pathogenesis of germ cell tumours. Histopathology. 2004;45(2):155ā€“61.

    PubMedĀ  CASĀ  Google ScholarĀ 

  254. Burke AP, Mostofi FK. Intratubular malignant germ cells in testicular biopsies: clinical course and identification by staining for placental alkaline phosphatase. Mod Pathol. 1988;1(6):475ā€“9.

    Google ScholarĀ 

  255. Rajpert-De Meyts E, Hanstein R, Jorgensen N, Graem N, Vogt PH, Skakkebaek NE. Developmental expression of POU5F1 (OCT-3/4) in normal and dysgenetic human gonads. Hum Reprod. 2004;19(6):1338ā€“44.

    Google ScholarĀ 

  256. Looijenga LH, Stoop H, de Leeuw HP, de Gouveia Brazao CA, Gillis AJ, van Roozendaal KE, et al. POU5F1 (OCT3/4) identifies cells with pluripotent potential in human germ cell tumors. Cancer Res. 2003;63(9):2244ā€“50.

    PubMedĀ  CASĀ  Google ScholarĀ 

  257. Jones TD, Ulbright TM, Eble JN, Cheng L. OCT4: a sensitive and specific biomarker for intratubular germ cell neoplasia of the testis. Clin Cancer Res. 2004;10(24):8544ā€“7.

    PubMedĀ  CASĀ  Google ScholarĀ 

  258. Ottesen AM, Skakkebaek NE, Lundsteen C, Leffers H, Larsen J, Rajpert-De Meyts E. High-resolution comparative genomic hybridization detects extra chromosome arm 12p material in most cases of carcinoma in situ adjacent to overt germ cell tumors, but not before the invasive tumor development. Genes Chromosomes Cancer. 2003;38(2):117ā€“25.

    PubMedĀ  CASĀ  Google ScholarĀ 

  259. Moch H, Cubilla AL, Humphrey PA, Reuter VE, Ulbright TM. The 2016 WHO classification of tumours of the urinary system and male genital organs-part a: renal, penile, and testicular tumours. Eur Urol. 2016;70(1):93ā€“105.

    PubMedĀ  Google ScholarĀ 

  260. Leibovitch I, Baniel J, Rowland RG, Smith ER Jr, Ludlow JK, Donohue JP. Malignant testicular neoplasms in immunosuppressed patients. J Urol. 1996;155(6):1938ā€“42.

    PubMedĀ  CASĀ  Google ScholarĀ 

  261. Halme A, Kellokumpu-Lehtinen P, Lehtonen T, Teppo L. Morphology of testicular germ cell tumours in treated and untreated cryptorchidism. Br J Urol. 1989;64(1):78ā€“83.

    PubMedĀ  CASĀ  Google ScholarĀ 

  262. Aparicio J, Garcia del Muro X, Maroto P, Paz-Ares L, Alba E, Saenz A, et al. Multicenter study evaluating a dual policy of postorchiectomy surveillance and selective adjuvant single-agent carboplatin for patients with clinical stage I seminoma. Ann Oncol. 2003;14(6):867ā€“72.

    PubMedĀ  CASĀ  Google ScholarĀ 

  263. Groll RJ, Warde P, Jewett MA. A comprehensive systematic review of testicular germ cell tumor surveillance. Crit Rev Oncol Hematol. 2007;64(3):182ā€“97.

    PubMedĀ  CASĀ  Google ScholarĀ 

  264. Oliver RT, Mason MD, Mead GM, von der Maase H, Rustin GJ, Joffe JK, et al. Radiotherapy versus single-dose carboplatin in adjuvant treatment of stage I seminoma: a randomised trial. Lancet. 2005;366(9482):293ā€“300.

    PubMedĀ  CASĀ  Google ScholarĀ 

  265. Oliver RT, Mead GM, Rustin GJ, Joffe JK, Aass N, Coleman R, et al. Randomized trial of carboplatin versus radiotherapy for stage I seminoma: mature results on relapse and contralateral testis cancer rates in MRC TE19/EORTC 30982 study (ISRCTN27163214). J Clin Oncol. 2011;29(8):957ā€“62.

    PubMedĀ  CASĀ  Google ScholarĀ 

  266. Motzer RJ, Jonasch E, Agarwal N, Beard C, Bhayani S, Bolger GB, et al. Testicular Cancer, version 2.2015. J Natl Compr Canc Netw. 2015;13(6):772ā€“99.

    PubMedĀ  Google ScholarĀ 

  267. Damjanov I, Osborn M, Miettinen M. Keratin 7 is a marker for a subset of trophoblastic cells in human germ cell tumors. Arch Pathol Lab Med. 1990;114(1):81ā€“3.

    PubMedĀ  CASĀ  Google ScholarĀ 

  268. Mostofi FK, Sesterhenn IA. Pathology of germ cell tumors of testes. Prog Clin Biol Res. 1985;203:1ā€“34.

    PubMedĀ  CASĀ  Google ScholarĀ 

  269. Weissbach L, Bussar-Maatz R, Lohrs U, Schubert GE, Mann K, Hartmann M, et al. Prognostic factors in seminomas with special respect to HCG: results of a prospective multicenter study. Seminoma Study Group. Eur Urol. 1999;36(6):601ā€“8.

    PubMedĀ  CASĀ  Google ScholarĀ 

  270. Al Nazer M, Al Dayel F. Tubular seminoma: case report and literature review. Ann Saudi Med. 2001;21(5ā€“6):334ā€“6.

    PubMedĀ  Google ScholarĀ 

  271. Young RH, Finlayson N, Scully RE. Tubular seminoma. Report of a case. Arch Pathol Lab Med. 1989;113(4):414ā€“6.

    PubMedĀ  CASĀ  Google ScholarĀ 

  272. Henley JD, Young RH, Wade CL, Ulbright TM. Seminomas with exclusive intertubular growth: a report of 12 clinically and grossly inconspicuous tumors. Am J Surg Pathol. 2004;28(9):1163ā€“8.

    PubMedĀ  Google ScholarĀ 

  273. Cheville JC, Rao S, Iczkowski KA, Lohse CM, Pankratz VS. Cytokeratin expression in seminoma of the human testis. Am J Clin Pathol. 2000;113(4):583ā€“8.

    PubMedĀ  CASĀ  Google ScholarĀ 

  274. Cutcutache I, Suzuki Y, Tan IB, Ramgopal S, Zhang S, Ramnarayanan K, et al. Exome-wide sequencing shows low mutation rates and identifies novel mutated genes in seminomas. Eur Urol. 2015;68(1):77ā€“83.

    PubMedĀ  CASĀ  Google ScholarĀ 

  275. McIntyre A, Summersgill B, Grygalewicz B, Gillis AJ, Stoop J, van Gurp RJ, et al. Amplification and overexpression of the KIT gene is associated with progression in the seminoma subtype of testicular germ cell tumors of adolescents and adults. Cancer Res. 2005;65(18):8085ā€“9.

    PubMedĀ  CASĀ  Google ScholarĀ 

  276. Jones RH, Vasey PA. Part I: testicular cancer--management of early disease. Lancet Oncol. 2003;4(12):730ā€“7.

    PubMedĀ  Google ScholarĀ 

  277. Albers P, Siener R, Kliesch S, Weissbach L, Krege S, Sparwasser C, et al. Risk factors for relapse in clinical stage I nonseminomatous testicular germ cell tumors: results of the German Testicular Cancer Study Group Trial. J Clin Oncol. 2003;21(8):1505ā€“12.

    PubMedĀ  Google ScholarĀ 

  278. Mostofi FK, Sesterhenn IA, Davis CJ Jr. Developments in histopathology of testicular germ cell tumors. Semin Urol. 1988;6(3):171ā€“88.

    PubMedĀ  CASĀ  Google ScholarĀ 

  279. Kao CS, Ulbright TM, Young RH, Idrees MT. Testicular embryonal carcinoma: a morphologic study of 180 cases highlighting unusual and unemphasized aspects. Am J Surg Pathol. 2014;38(5):689ā€“97.

    PubMedĀ  Google ScholarĀ 

  280. Rodriguez PN, Hafez GR, Messing EM. Nonseminomatous germ cell tumor of the testicle: does extensive staging of the primary tumor predict the likelihood of metastatic disease? J Urol. 1986;136(3):604ā€“8.

    PubMedĀ  CASĀ  Google ScholarĀ 

  281. Ulbright TM, Tickoo SK, Berney DM, Srigley JR. Members of the IIiDUPG. Best practices recommendations in the application of immunohistochemistry in testicular tumors: report from the International Society of Urological Pathology consensus conference. Am J Surg Pathol. 2014;38(8):e50ā€“9.

    PubMedĀ  Google ScholarĀ 

  282. van Echten J, Oosterhuis JW, Looijenga LH, van de Pol M, Wiersema J, te Meerman GJ, et al. No recurrent structural abnormalities apart from i(12p) in primary germ cell tumors of the adult testis. Genes Chromosomes Cancer. 1995;14(2):133ā€“44.

    PubMedĀ  Google ScholarĀ 

  283. Stang A, Trabert B, Wentzensen N, Cook MB, Rusner C, Oosterhuis JW, et al. Gonadal and extragonadal germ cell tumours in the United States, 1973-2007. Int J Androl. 2012;35(4):616ā€“25.

    PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  284. Cornejo KM, Frazier L, Lee RS, Kozakewich HP, Young RH. Yolk sac tumor of the testis in infants and children: a clinicopathologic analysis of 33 cases. Am J Surg Pathol. 2015;39(8):1121ā€“31.

    PubMedĀ  Google ScholarĀ 

  285. Hawkins E, Heifetz SA, Giller R, Cushing B. The prepubertal testis (prenatal and postnatal): its relationship to intratubular germ cell neoplasia: a combined Pediatric Oncology Group and Childrenā€™s Cancer Study Group. Hum Pathol. 1997;28(4):404ā€“10.

    PubMedĀ  CASĀ  Google ScholarĀ 

  286. Perlman EJ, Hu J, Ho D, Cushing B, Lauer S, Castleberry RP. Genetic analysis of childhood endodermal sinus tumors by comparative genomic hybridization. J Pediatr Hematol Oncol. 2000;22(2):100ā€“5.

    PubMedĀ  CASĀ  Google ScholarĀ 

  287. Rogers PC, Olson TA, Cullen JW, Billmire DF, Marina N, Rescorla F, et al. Treatment of children and adolescents with stage II testicular and stages I and II ovarian malignant germ cell tumors: a Pediatric Intergroup Study--Pediatric Oncology Group 9048 and Childrenā€™s Cancer Group 8891. J Clin Oncol. 2004;22(17):3563ā€“9.

    PubMedĀ  Google ScholarĀ 

  288. Krag Jacobsen G, Barlebo H, Olsen J, Schultz HP, Starklint H, Sogaard H, et al. Testicular germ cell tumours in Denmark 1976-1980. Pathology of 1058 consecutive cases. Acta Radiol Oncol. 1984;23(4):239ā€“47.

    PubMedĀ  CASĀ  Google ScholarĀ 

  289. von Hochstetter AR, Hedinger CE. The differential diagnosis of testicular germ cell tumors in theory and practice. A critical analysis of two major systems of classification and review of 389 cases. Virchows Arch A Pathol Anat Histol. 1982;396(3):247ā€“77.

    Google ScholarĀ 

  290. Freedman LS, Parkinson MC, Jones WG, Oliver RT, Peckham MJ, Read G, et al. Histopathology in the prediction of relapse of patients with stage I testicular teratoma treated by orchidectomy alone. Lancet. 1987;2(8554):294ā€“8.

    PubMedĀ  CASĀ  Google ScholarĀ 

  291. Logothetis CJ, Samuels ML, Trindade A, Grant C, Gomez L, Ayala A. The prognostic significance of endodermal sinus tumor histology among patients treated for stage III nonseminomatous germ cell tumors of the testes. Cancer. 1984;53(1):122ā€“8.

    PubMedĀ  CASĀ  Google ScholarĀ 

  292. Teilum G. Endodermal sinus tumors of the ovary and testis. Comparative morphogenesis of the so-called mesoephroma ovarii (Schiller) and extraembryonic (yolk sac-allantoic) structures of the rat's placenta. Cancer. 1959;12:1092ā€“105.

    PubMedĀ  CASĀ  Google ScholarĀ 

  293. Cao D, Li J, Guo CC, Allan RW, Humphrey PA. SALL4 is a novel diagnostic marker for testicular germ cell tumors. Am J Surg Pathol. 2009;33(7):1065ā€“77.

    PubMedĀ  Google ScholarĀ 

  294. Cao D, Humphrey PA, Allan RW. SALL4 is a novel sensitive and specific marker for metastatic germ cell tumors, with particular utility in detection of metastatic yolk sac tumors. Cancer. 2009;115(12):2640ā€“51.

    PubMedĀ  CASĀ  Google ScholarĀ 

  295. Ota S, Hishinuma M, Yamauchi N, Goto A, Morikawa T, Fujimura T, et al. Oncofetal protein glypican-3 in testicular germ-cell tumor. Virchows Arch. 2006;449(3):308ā€“14.

    PubMedĀ  CASĀ  Google ScholarĀ 

  296. Zynger DL, Dimov ND, Luan C, Teh BT, Yang XJ. Glypican 3: a novel marker in testicular germ cell tumors. Am J Surg Pathol. 2006;30(12):1570ā€“5.

    PubMedĀ  Google ScholarĀ 

  297. Talerman A, Haije WG, Baggerman L. Serum alphafetoprotein (AFP) in patients with germ cell tumors of the gonads and extragonadal sites: correlation between endodermal sinus (yolk sac) tumor and raised serum AFP. Cancer. 1980;46(2):380ā€“5.

    PubMedĀ  CASĀ  Google ScholarĀ 

  298. Manivel JC, Reinberg Y, Niehans GA, Fraley EE. Intratubular germ cell neoplasia in testicular teratomas and epidermoid cysts. Correlation with prognosis and possible biologic significance. Cancer. 1989;64(3):715ā€“20.

    PubMedĀ  CASĀ  Google ScholarĀ 

  299. Silver SA, Wiley JM, Perlman EJ. DNA ploidy analysis of pediatric germ cell tumors. Mod Pathol. 1994;7(9):951ā€“6.

    PubMedĀ  CASĀ  Google ScholarĀ 

  300. Semjen D, Kalman E, Tornoczky T, Szuhai K. Further evidence of the existence of benign teratomas of the postpubertal testis. Am J Surg Pathol. 2014;38(4):580ā€“1.

    PubMedĀ  Google ScholarĀ 

  301. Zhang C, Berney DM, Hirsch MS, Cheng L, Ulbright TM. Evidence supporting the existence of benign teratomas of the postpubertal testis: a clinical, histopathologic, and molecular genetic analysis of 25 cases. Am J Surg Pathol. 2013;37(6):827ā€“35.

    PubMedĀ  Google ScholarĀ 

  302. Dieckmann KP, Loy V. Epidermoid cyst of the testis: a review of clinical and histogenetic considerations. Br J Urol. 1994;73(4):436ā€“41.

    PubMedĀ  CASĀ  Google ScholarĀ 

  303. Shah KH, Maxted WC, Chun B. Epidermoid cysts of the testis: a report of three cases and an analysis of 141 cases from the world literature. Cancer. 1981;47(3):577ā€“82.

    PubMedĀ  CASĀ  Google ScholarĀ 

  304. Ulbright TM, Srigley JR. Dermoid cyst of the testis: a study of five postpubertal cases, including a pilomatrixoma-like variant, with evidence supporting its separate classification from mature testicular teratoma. Am J Surg Pathol. 2001;25(6):788ā€“93.

    PubMedĀ  CASĀ  Google ScholarĀ 

  305. Rushton HG, Belman AB, Sesterhenn I, Patterson K, Mostofi FK. Testicular sparing surgery for prepubertal teratoma of the testis: a clinical and pathological study. J Urol. 1990;144(3):726ā€“30.

    PubMedĀ  CASĀ  Google ScholarĀ 

  306. Kernek KM, Ulbright TM, Zhang S, Billings SD, Cummings OW, Henley JD, et al. Identical allelic losses in mature teratoma and other histologic components of malignant mixed germ cell tumors of the testis. Am J Pathol. 2003;163(6):2477ā€“84.

    PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  307. Leibovitch I, Foster RS, Ulbright TM, Donohue JP. Adult primary pure teratoma of the testis. The Indiana experience. Cancer. 1995;75(9):2244ā€“50.

    PubMedĀ  CASĀ  Google ScholarĀ 

  308. Foster RS, Baniel J, Leibovitch I, Curran M, Bihrle R, Rowland R, et al. Teratoma in the orchiectomy specimen and volume of metastasis are predictors of retroperitoneal teratoma in low stage nonseminomatous testis cancer. J Urol. 1996;155(6):1943ā€“5.

    PubMedĀ  CASĀ  Google ScholarĀ 

  309. Rabbani F, Gleave ME, Coppin CM, Murray N, Sullivan LD. Teratoma in primary testis tumor reduces complete response rates in the retroperitoneum after primary chemotherapy. The case for primary retroperitoneal lymph node dissection of stage IIb germ cell tumors with teratomatous elements. Cancer. 1996;78(3):480ā€“6.

    PubMedĀ  CASĀ  Google ScholarĀ 

  310. Alvarado-Cabrero I, Hernandez-Toriz N, Paner GP. Clinicopathologic analysis of choriocarcinoma as a pure or predominant component of germ cell tumor of the testis. Am J Surg Pathol. 2014;38(1):111ā€“8.

    PubMedĀ  Google ScholarĀ 

  311. Papiani G, Einhorn LH. Salvage chemotherapy with high-dose carboplatin plus etoposide and autologous peripheral blood stem cell transplant in male pure choriocarcinoma: a retrospective analysis of 13 cases. Bone Marrow Transplant. 2007;40(3):235ā€“7.

    PubMedĀ  CASĀ  Google ScholarĀ 

  312. Jensen JL, Venner PM. Predictive factors for outcome in treatment of metastatic nonseminomatous germ cell tumors. Urology. 1992;39(3):237ā€“42.

    PubMedĀ  CASĀ  Google ScholarĀ 

  313. Ulbright TM, Henley JD, Cummings OW, Foster RS, Cheng L. Cystic trophoblastic tumor: a nonaggressive lesion in postchemotherapy resections of patients with testicular germ cell tumors. Am J Surg Pathol. 2004;28(9):1212ā€“6.

    PubMedĀ  Google ScholarĀ 

  314. Berney DM, Shamash J, Pieroni K, Oliver RT. Loss of CD30 expression in metastatic embryonal carcinoma: the effects of chemotherapy? Histopathology. 2001;39(4):382ā€“5.

    PubMedĀ  CASĀ  Google ScholarĀ 

  315. Sung MT, Jones TD, Beck SD, Foster RS, Cheng L. OCT4 is superior to CD30 in the diagnosis of metastatic embryonal carcinomas after chemotherapy. Hum Pathol. 2006;37(6):662ā€“7.

    PubMedĀ  CASĀ  Google ScholarĀ 

  316. Miettinen M, Wang Z, McCue PA, Sarlomo-Rikala M, Rys J, Biernat W, et al. SALL4 expression in germ cell and non-germ cell tumors: a systematic immunohistochemical study of 3215 cases. Am J Surg Pathol. 2014;38(3):410ā€“20.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  317. Ulbright TM, Loehrer PJ, Roth LM, Einhorn LH, Williams SD, Clark SA. The development of non-germ cell malignancies within germ cell tumors. A clinicopathologic study of 11 cases. Cancer. 1984;54(9):1824ā€“33.

    PubMedĀ  CASĀ  Google ScholarĀ 

  318. Magers MJ, Kao CS, Cole CD, Rice KR, Foster RS, Einhorn LH, et al. ā€œSomatic-typeā€ malignancies arising from testicular germ cell tumors: a clinicopathologic study of 124 cases with emphasis on glandular tumors supporting frequent yolk sac tumor origin. Am J Surg Pathol. 2014;38(10):1396ā€“409.

    PubMedĀ  Google ScholarĀ 

  319. Rice KR, Magers MJ, Beck SD, Cary KC, Einhorn LH, Ulbright TM, et al. Management of germ cell tumors with somatic type malignancy: pathological features, prognostic factors and survival outcomes. J Urol. 2014;192(5):1403ā€“9.

    PubMedĀ  Google ScholarĀ 

  320. True LD, Otis CN, Rosai J, Scully RE, Delprado W. Spermatocytic seminoma of testis with sarcomatous transformation. Am J Surg Pathol. 1988;12(10):806.

    PubMedĀ  CASĀ  Google ScholarĀ 

  321. Guo CC, Punar M, Contreras AL, Tu SM, Pisters L, Tamboli P, et al. Testicular germ cell tumors with sarcomatous components: an analysis of 33 cases. Am J Surg Pathol. 2009;33(8):1173ā€“8.

    PubMedĀ  Google ScholarĀ 

  322. Malagon HD, Valdez AM, Moran CA, Suster S. Germ cell tumors with sarcomatous components: a clinicopathologic and immunohistochemical study of 46 cases. Am J Surg Pathol. 2007;31(9):1356ā€“62.

    PubMedĀ  Google ScholarĀ 

  323. Michael H, Hull MT, Ulbright TM, Foster RS, Miller KD. Primitive neuroectodermal tumors arising in testicular germ cell neoplasms. Am J Surg Pathol. 1997;21(8):896ā€“904.

    PubMedĀ  CASĀ  Google ScholarĀ 

  324. Ulbright TM, Hattab EM, Zhang S, Ehrlich Y, Foster RS, Einhorn LH, et al. Primitive neuroectodermal tumors in patients with testicular germ cell tumors usually resemble pediatric-type central nervous system embryonal neoplasms and lack chromosome 22 rearrangements. Mod Pathol. 2010;23(7):972ā€“80.

    PubMedĀ  CASĀ  Google ScholarĀ 

  325. Oldenburg J, Martin JM, Fossa SD. Late relapses of germ cell malignancies: incidence, management, and prognosis. J Clin Oncol. 2006;24(35):5503ā€“11.

    PubMedĀ  Google ScholarĀ 

  326. Baniel J, Foster RS, Gonin R, Messemer JE, Donohue JP, Einhorn LH. Late relapse of testicular cancer. J Clin Oncol. 1995;13(5):1170ā€“6.

    PubMedĀ  CASĀ  Google ScholarĀ 

  327. Sharp DS, Carver BS, Eggener SE, Kondagunta GV, Motzer RJ, Bosl GJ, et al. Clinical outcome and predictors of survival in late relapse of germ cell tumor. J Clin Oncol. 2008;26(34):5524ā€“9.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  328. Geldart TR, Gale J, McKendrick J, Kirby J, Mead G. Late relapse of metastatic testicular nonseminomatous germ cell cancer: surgery is needed for cure. BJU Int. 2006;98(2):353ā€“8.

    PubMedĀ  Google ScholarĀ 

  329. Michael H, Lucia J, Foster RS, Ulbright TM. The pathology of late recurrence of testicular germ cell tumors. Am J Surg Pathol. 2000;24(2):257ā€“73.

    PubMedĀ  CASĀ  Google ScholarĀ 

  330. Jou P, Maclennan GT. Leydig cell tumor of the testis. J Urol. 2009;181(5):2299ā€“300.

    PubMedĀ  Google ScholarĀ 

  331. Talon I, Moog R, Kauffmann I, Grandadam S, Becmeur F. Sertoli cell tumor of the testis in children: reevaluation of a rarely encountered tumor. J Pediatr Hematol Oncol. 2005;27(9):491ā€“4.

    PubMedĀ  Google ScholarĀ 

  332. Adayener C, Akyol I, Sen B, Ates F, Haholu A, Soydan H, et al. Sertoli cell tumor of the testis: a case with late metastasis. Int Urol Nephrol. 2008;40(4):1005ā€“8.

    PubMedĀ  Google ScholarĀ 

  333. Young RH, Koelliker DD, Scully RE. Sertoli cell tumors of the testis, not otherwise specified: a clinicopathologic analysis of 60 cases. Am J Surg Pathol. 1998;22(6):709ā€“21.

    PubMedĀ  CASĀ  Google ScholarĀ 

  334. Henley JD, Young RH, Ulbright TM. Malignant Sertoli cell tumors of the testis: a study of 13 examples of a neoplasm frequently misinterpreted as seminoma. Am J Surg Pathol. 2002;26(5):541ā€“50.

    PubMedĀ  Google ScholarĀ 

  335. Halat SK, Ponsky LE, MacLennan GT. Large cell calcifying Sertoli cell tumor of testis. J Urol. 2007;177(6):2338.

    PubMedĀ  Google ScholarĀ 

  336. Ulbright TM, Amin MB, Young RH. Intratubular large cell hyalinizing sertoli cell neoplasia of the testis: a report of 8 cases of a distinctive lesion of the Peutz-Jeghers syndrome. Am J Surg Pathol. 2007;31(6):827ā€“35.

    PubMedĀ  Google ScholarĀ 

  337. Nogales FF, Andujar M, Zuluaga A, Garcia-Puche JL. Malignant large cell calcifying Sertoli cell tumor of the testis. J Urol. 1995;153(6):1935ā€“7.

    PubMedĀ  CASĀ  Google ScholarĀ 

  338. Tetu B, Ro JY, Ayala AG. Large cell calcifying Sertoli cell tumor of the testis. A clinicopathologic, immunohistochemical, and ultrastructural study of two cases. Am J Clin Pathol. 1991;96(6):717ā€“22.

    PubMedĀ  CASĀ  Google ScholarĀ 

  339. Kratzer SS, Ulbright TM, Talerman A, Srigley JR, Roth LM, Wahle GR, et al. Large cell calcifying Sertoli cell tumor of the testis: contrasting features of six malignant and six benign tumors and a review of the literature. Am J Surg Pathol. 1997;21(11):1271ā€“80.

    PubMedĀ  CASĀ  Google ScholarĀ 

  340. Kao CS, Kum JB, Idrees MT, Ulbright TM. Sclerosing Sertoli cell tumor of the testis: a clinicopathologic study of 20 cases. Am J Surg Pathol. 2014;38(4):510ā€“7.

    PubMedĀ  Google ScholarĀ 

  341. Colecchia M. Observations on the paper ā€œsclerosing Sertoli cell tumor of the testis: a clinicopathologic study of 20 casesā€ by Kao et al. Am J Surg Pathol. 2014;38(8):1160.

    PubMedĀ  Google ScholarĀ 

  342. Esber CM, Shabsigh A, Zynger DL. Sclerosing Sertoli cell tumor without expression of typical sex cord stromal tumor markers: case report and literature review. Pathol Res Pract. 2012;208(2):121ā€“5.

    PubMedĀ  CASĀ  Google ScholarĀ 

  343. Schubert TE, Stoehr R, Hartmann A, Schone S, Lobelenz M, Mikuz G. Adult type granulosa cell tumor of the testis with a heterologous sarcomatous component: case report and review of the literature. Diagn Pathol. 2014;9:107.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  344. Mohapatra A, Potretzke AM, Knight BA, Han M, Figenshau RS. Metastatic Granulosa cell tumor of the testis: clinical presentation and management. Case Rep Urol. 2016;2016:9016728.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  345. Harrison MR, Huang W, Liu G, Gee J. Response to antiangiogenesis therapy in a patient with advanced adult-type testicular granulosa cell tumor. Oncology (Williston Park, NY). 2009;23(9):792ā€“5.

    Google ScholarĀ 

  346. Gohji K, Higuchi A, Fujii A, Kizaki T. Malignant gonadal stromal tumor. Urology. 1994;43(2):244ā€“7.

    PubMedĀ  CASĀ  Google ScholarĀ 

  347. Cornejo KM, Young RH. Adult granulosa cell tumors of the testis: a report of 32 cases. Am J Surg Pathol. 2014;38(9):1242ā€“50.

    PubMedĀ  Google ScholarĀ 

  348. Kao CS, Cornejo KM, Ulbright TM, Young RH. Juvenile granulosa cell tumors of the testis: a clinicopathologic study of 70 cases with emphasis on its wide morphologic spectrum. Am J Surg Pathol. 2015;39(9):1159ā€“69.

    PubMedĀ  Google ScholarĀ 

  349. May D, Shamberger R, Newbury R, Teele RL. Juvenile granulosa cell tumor of an intraabdominal testis. Pediatr Radiol. 1992;22(7):507ā€“8.

    PubMedĀ  CASĀ  Google ScholarĀ 

  350. Young RH, Lawrence WD, Scully RE. Juvenile granulosa cell tumor--another neoplasm associated with abnormal chromosomes and ambiguous genitalia. A report of three cases. Am J Surg Pathol. 1985;9(10):737ā€“43.

    PubMedĀ  CASĀ  Google ScholarĀ 

  351. Shukla AR, Huff DS, Canning DA, Filmer RB, Snyder HM 3rd, Carpintieri D, et al. Juvenile granulosa cell tumor of the testis: contemporary clinical management and pathological diagnosis. J Urol. 2004;171(5):1900ā€“2.

    PubMedĀ  Google ScholarĀ 

  352. Lawrence WD, Young RH, Scully RE. Juvenile granulosa cell tumor of the infantile testis. A report of 14 cases. Am J Surg Pathol. 1985;9(2):87ā€“94.

    PubMedĀ  CASĀ  Google ScholarĀ 

  353. Groisman GM, Dische MR, Fine EM, Unger PD. Juvenile granulosa cell tumor of the testis: a comparative immunohistochemical study with normal infantile gonads. Pediatr Pathol. 1993;13(4):389ā€“400.

    PubMedĀ  CASĀ  Google ScholarĀ 

  354. Zhang M, Kao CS, Ulbright TM, Epstein JI. Testicular fibrothecoma: a morphologic and immunohistochemical study of 16 cases. Am J Surg Pathol. 2013;37(8):1208ā€“14.

    PubMedĀ  Google ScholarĀ 

  355. Jones MA, Young RH, Scully RE. Benign fibromatous tumors of the testis and paratesticular region: a report of 9 cases with a proposed classification of fibromatous tumors and tumor-like lesions. Am J Surg Pathol. 1997;21(3):296ā€“305.

    PubMedĀ  CASĀ  Google ScholarĀ 

  356. Scully RE. Gonadoblastoma. A review of 74 cases. Cancer. 1970;25(6):1340ā€“56.

    PubMedĀ  CASĀ  Google ScholarĀ 

  357. Scully RE. Gonadoblastoma; a gonadal tumor related to the dysgerminoma (seminoma) and capable of sex-hormone production. Cancer. 1953;6(3):455ā€“63.

    PubMedĀ  CASĀ  Google ScholarĀ 

  358. Dendrinos ML, Smorgick N, Marsh CA, Smith YR, Quint EH. Occurrence of gonadoblastoma in patients with 45,X/46,XY mosaicism. J Pediatr Adolesc Gynecol. 2015;28(3):192ā€“5.

    PubMedĀ  Google ScholarĀ 

  359. Ng SB, Yong MH, Knight LA, Lee VK, Nadarajah S, Stoop H, et al. Gonadoblastoma-associated mixed germ cell tumour in 46,XY complete gonadal dysgenesis (Swyer syndrome): analysis of Y chromosomal genotype and OCT3/4 and TSPY expression profile. Histopathology. 2008;52(5):644ā€“6.

    PubMedĀ  CASĀ  Google ScholarĀ 

  360. Cools M, Stoop H, Kersemaekers AM, Drop SL, Wolffenbuttel KP, Bourguignon JP, et al. Gonadoblastoma arising in undifferentiated gonadal tissue within dysgenetic gonads. J Clin Endocrinol Metab. 2006;91(6):2404ā€“13.

    PubMedĀ  CASĀ  Google ScholarĀ 

  361. Kersemaekers AM, Honecker F, Stoop H, Cools M, Molier M, Wolffenbuttel K, et al. Identification of germ cells at risk for neoplastic transformation in gonadoblastoma: an immunohistochemical study for OCT3/4 and TSPY. Hum Pathol. 2005;36(5):512ā€“21.

    PubMedĀ  CASĀ  Google ScholarĀ 

  362. Kao CS, Ulbright TM, Idrees MT. Gonadoblastoma: an immunohistochemical study and comparison to Sertoli cell nodule with intratubular germ cell neoplasia, with pathogenetic implications. Histopathology. 2014;65(6):861ā€“7.

    PubMedĀ  Google ScholarĀ 

  363. Tarjan M, Sarkissov G, Tot T. Unclassified sex cord/gonadal stromal testis tumor with predominance of spindle cells. APMIS. 2006;114(6):465ā€“9.

    PubMedĀ  CASĀ  Google ScholarĀ 

  364. Dieckmann KP, Loy V. Response of metastasized sex cord gonadal stromal tumor of the testis to cisplatin-based chemotherapy. J Urol. 1994;151(4):1024ā€“6.

    PubMedĀ  CASĀ  Google ScholarĀ 

  365. Renshaw AA, Gordon M, Corless CL. Immunohistochemistry of unclassified sex cord-stromal tumors of the testis with a predominance of spindle cells. Mod Pathol. 1997;10(7):693ā€“700.

    PubMedĀ  CASĀ  Google ScholarĀ 

  366. Ulbright TM, Srigley JR, Reuter VE, Wojno K, Roth LM, Young RH. Sex cord-stromal tumors of the testis with entrapped germ cells: a lesion mimicking unclassified mixed germ cell sex cord-stromal tumors. Am J Surg Pathol. 2000;24(4):535ā€“42.

    PubMedĀ  CASĀ  Google ScholarĀ 

  367. Burger T, Schildhaus HU, Inniger R, Hansen J, Mayer P, Schweyer S, et al. Ovarian-type epithelial tumours of the testis: immunohistochemical and molecular analysis of two serous borderline tumours of the testis. Diagn Pathol. 2015;10:118.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  368. Scholz B, Beckert M, Mordstein V, Hohmann N, Walther R, Papadopoulos T. Seromucinous borderline tumor of the testis-a case report. Hum Pathol. 2017;60:188ā€“91.

    PubMedĀ  Google ScholarĀ 

  369. Tulunay O, Gogus C, Baltaci S, Bulut S. Clear cell adenocarcinoma of the tunica vaginalis of the testis with an adjacent uterus-like tissue. Pathol Int. 2004;54(8):641ā€“7.

    PubMedĀ  Google ScholarĀ 

  370. Blumberg HM, Hendrix LE. Serous papillary adenocarcinoma of the tunica vaginalis of the testis with metastasis. Cancer. 1991;67(5):1450ā€“3.

    PubMedĀ  CASĀ  Google ScholarĀ 

  371. Aguirre P, Thor AD, Scully RE. Ovarian endometrioid carcinomas resembling sex cord-stromal tumors. An immunohistochemical study. Int J Gynecol Pathol. 1989;8(4):364ā€“73.

    PubMedĀ  CASĀ  Google ScholarĀ 

  372. Young RH, Scully RE. Testicular and paratesticular tumors and tumor-like lesions of ovarian common epithelial and mullerian types. A report of four cases and review of the literature. Am J Clin Pathol. 1986;86(2):146ā€“52.

    PubMedĀ  CASĀ  Google ScholarĀ 

  373. Velazquez EF, Barreto JE, Cubilla AL. Penis and distal urethra. In: Mills SE, editor. Histology for pathologists. Philadelphia: LWW; 2012. p. 1027ā€“44.

    Google ScholarĀ 

  374. Sadler TW. Langmanā€™s medical embryology. Baltimore: LWW; 2012.

    Google ScholarĀ 

  375. Schoenwolf GC, Bleyl SB, Brauer PR, et al. Larsenā€™s human embryology. Pholadelphia: Elsevier; 2015.

    Google ScholarĀ 

  376. Cubilla AL, Piris A, Pfannl R, et al. Anatomic levels: important landmarks in penectomy specimens: a detailed anatomic and histologic study based on examination of 44 cases. Am J Surg Pathol. 2001;25:1091ā€“4.

    PubMedĀ  CASĀ  Google ScholarĀ 

  377. Epstein JI, Cubilla AL, Humphrey PA. Tumors of the prostate gland, seminal vesicles, penis, and scrotum. Washington, DC: American Registry of Pathology and AFIP; 2011.

    Google ScholarĀ 

  378. Velazquez EF, Bock A, Soskin A, et al. Preputial variability and preferential association of long phimotic foreskins with penile cancer: an anatomic comparative study of types of foreskin in a general population and cancer patients. Am J Surg Pathol. 2003;27:994ā€“8.

    PubMedĀ  Google ScholarĀ 

  379. Velazquez EF, Amin MB, Epstein JI, et al. Protocol for the examination of specimens from patients with carcinoma of the penis. 2013. Available at: www.cap.org/cancerprotocols. Accessed 12/20/2016, 2016.

  380. Hadway P, Corbishley CM, Watkin NA. Total glans resurfacing for premalignant lesions of the penis: initial outcome data. BJU Int. 2006;98:532ā€“6.

    PubMedĀ  Google ScholarĀ 

  381. Shabbir M, Muneer A, Kalsi J, et al. Glans resurfacing for the treatment of carcinoma in situ of the penis: surgical technique and outcomes. Eur Urol. 2011;59:142ā€“7.

    PubMedĀ  Google ScholarĀ 

  382. Corbishley CM, Tinwell B, Kaul A, et al. Glans resurfacing for precancerous and superficially invasive carcinomas of the glans penis: pathological specimen handling and reporting. Semin Diagn Pathol. 2015;32:232ā€“7.

    PubMedĀ  Google ScholarĀ 

  383. Moch H, Humphrey PA, Ulbright TM, et al. WHO classification of tumours of the urinary system and male genital organs. 4th ed. Lyon: IARC Press; 2016.

    Google ScholarĀ 

  384. Chaux A, Velazquez EF, Amin A, et al. Distribution and characterization of subtypes of penile intraepithelial neoplasia and their association with invasive carcinomas: a pathological study of 139 lesions in 121 patients. Hum Pathol. 2012;43:1020ā€“7.

    PubMedĀ  Google ScholarĀ 

  385. Oertell J, Caballero C, Iglesias M, et al. Differentiated precursor lesions and low-grade variants of squamous cell carcinomas are frequent findings in foreskins of patients from a region of high penile cancer incidence. Histopathology. 2011;58:925ā€“33.

    PubMedĀ  Google ScholarĀ 

  386. Chaux A, Pfannl R, Lloveras B, et al. Distinctive association of p16INK4a overexpression with penile intraepithelial neoplasia depicting warty and/or basaloid features: a study of 141 cases evaluating a new nomenclature. Am J Surg Pathol. 2010;34:385ā€“92.

    PubMedĀ  Google ScholarĀ 

  387. Cubilla AL, Velazquez EF, Young RH. Epithelial lesions associated with invasive penile squamous cell carcinoma: a pathologic study of 288 cases. Int J Surg Pathol. 2004;12:351ā€“64.

    PubMedĀ  Google ScholarĀ 

  388. Kurman RJ, Toki T, Schiffman MH. Basaloid and warty carcinomas of the vulva. Distinctive types of squamous cell carcinoma frequently associated with human papillomaviruses. Am J Surg Pathol. 1993;17:133ā€“45.

    PubMedĀ  CASĀ  Google ScholarĀ 

  389. Gillison ML, Koch WM, Capone RB, et al. Evidence for a causal association between human papillomavirus and a subset of head and neck cancers. J Natl Cancer Inst. 2000;92:709ā€“20.

    PubMedĀ  CASĀ  Google ScholarĀ 

  390. Lin BM, Wang H, Dā€™Souza G, et al. Long-term prognosis and risk factors among patients with HPV-associated oropharyngeal squamous cell carcinoma. Cancer. 2013;119:3462ā€“71.

    PubMedĀ  Google ScholarĀ 

  391. Westra WH. Detection of human papillomavirus (HPV) in clinical samples: evolving methods and strategies for the accurate determination of HPV status of head and neck carcinomas. Oral Oncol. 2014;50:771ā€“9.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  392. Gregoire L, Cubilla AL, Reuter VE, et al. Preferential association of human papillomavirus with high-grade histologic variants of penile-invasive squamous cell carcinoma. J Natl Cancer Inst. 1995;87:1705ā€“9.

    PubMedĀ  CASĀ  Google ScholarĀ 

  393. Rubin MA, Kleter B, Zhou M, et al. Detection and typing of human papillomavirus DNA in penile carcinoma: evidence for multiple independent pathways of penile carcinogenesis. Am J Pathol. 2001;159:1211ā€“8.

    PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  394. Cubilla AL, Lloveras B, Alejo M, et al. The basaloid cell is the best tissue marker for human papillomavirus in invasive penile squamous cell carcinoma: a study of 202 cases from Paraguay. Am J Surg Pathol. 2010;34:104ā€“14.

    PubMedĀ  Google ScholarĀ 

  395. Alemany L, Cubilla A, Halec G, et al. Role of human papillomavirus in penile carcinomas worldwide. Eur Urol. 2016;69:953ā€“61.

    PubMedĀ  Google ScholarĀ 

  396. Sanchez DF, Canete S, Fernandez-Nestosa MJ, et al. HPV- and non-HPV-related subtypes of penile squamous cell carcinoma (SCC): morphological features and differential diagnosis according to the new WHO classification (2015). Semin Diagn Pathol. 2015;32:198ā€“221.

    PubMedĀ  Google ScholarĀ 

  397. Cubilla AL, Barreto J, Caballero C, et al. Pathologic features of epidermoid carcinoma of the penis. A prospective study of 66 cases. Am J Surg Pathol. 1993;17:753ā€“63.

    PubMedĀ  CASĀ  Google ScholarĀ 

  398. Chaux A, Han JS, Lee S, et al. Immunohistochemical profile of the penile urethra and differential expression of GATA3 in urothelial versus squamous cell carcinomas of the penile urethra. Hum Pathol. 2013;44:2760ā€“7.

    PubMedĀ  CASĀ  Google ScholarĀ 

  399. Crapanzano JP, Loukeris K, Borczuk AC, et al. Cytological, histological, and immunohistochemical findings of pulmonary carcinomas with basaloid features. Diagn Cytopathol. 2011;39:92ā€“100.

    PubMedĀ  Google ScholarĀ 

  400. Cubilla AL, Lloveras B, Alejo M, et al. Value of p16(INK)(4)(a) in the pathology of invasive penile squamous cell carcinomas: a report of 202 cases. Am J Surg Pathol. 2011;35:253ā€“61.

    PubMedĀ  Google ScholarĀ 

  401. CaƱete-Portillo S, Clavero O, Sanchez DF, et al. Medullary carcinoma of the penis. A distinctive HPV-related neoplasm: a report of 12 cases. Am J Surg Pathol. 2017;41(4):535ā€“40.

    Google ScholarĀ 

  402. Cubilla AL, Reuter V, Velazquez E, et al. Histologic classification of penile carcinoma and its relation to outcome in 61 patients with primary resection. Int J Surg Pathol. 2001;9:111ā€“20.

    PubMedĀ  CASĀ  Google ScholarĀ 

  403. Guimaraes GC, Cunha IW, Soares FA, et al. Penile squamous cell carcinoma clinicopathological features, nodal metastasis and outcome in 333 cases. J Urol. 2009;182:528ā€“34. discussion 534.

    PubMedĀ  Google ScholarĀ 

  404. Sanchez DF, Soares F, Alvarado-Cabrero I, et al. Pathological factors, behavior, and histological prognostic risk groups in subtypes of penile squamous cell carcinomas (SCC). Semin Diagn Pathol. 2015;32:222ā€“31.

    PubMedĀ  Google ScholarĀ 

  405. Ackerman LV. Verrucous carcinoma of the oral cavity. Surgery. 1948;23:670ā€“8.

    PubMedĀ  CASĀ  Google ScholarĀ 

  406. Chaux A, Reuter V, Lezcano C, et al. Comparison of morphologic features and outcome of resected recurrent and nonrecurrent squamous cell carcinoma of the penis: a study of 81 cases. Am J Surg Pathol. 2009;33:1299ā€“306.

    PubMedĀ  Google ScholarĀ 

  407. Kato N, Onozuka T, Yasukawa K, et al. Penile hybrid verrucous-squamous carcinoma associated with a superficial inguinal lymph node metastasis. Am J Dermatopathol. 2000;22:339ā€“43.

    PubMedĀ  CASĀ  Google ScholarĀ 

  408. Barreto JE, Velazquez EF, Ayala E, et al. Carcinoma cuniculatum: a distinctive variant of penile squamous cell carcinoma: report of 7 cases. Am J Surg Pathol. 2007;31:71ā€“5.

    PubMedĀ  Google ScholarĀ 

  409. Chaux A, Soares F, Rodriguez I, et al. Papillary squamous cell carcinoma, not otherwise specified (NOS) of the penis: clinicopathologic features, differential diagnosis, and outcome of 35 cases. Am J Surg Pathol. 2010;34:223ā€“30.

    PubMedĀ  Google ScholarĀ 

  410. Cubilla AL, Velazques EF, Reuter VE, et al. Warty (condylomatous) squamous cell carcinoma of the penis: a report of 11 cases and proposed classification of ā€˜verruciformā€™ penile tumors. Am J Surg Pathol. 2000;24:505ā€“12.

    PubMedĀ  CASĀ  Google ScholarĀ 

  411. Cunha IW, Guimaraes GC, Soares F, et al. Pseudoglandular (adenoid, acantholytic) penile squamous cell carcinoma: a clinicopathologic and outcome study of 7 patients. Am J Surg Pathol. 2009;33:551ā€“5.

    PubMedĀ  Google ScholarĀ 

  412. Alvarado-Cabrero I, Sanchez DF, Piedras D, et al. The variable morphological spectrum of penile basaloid carcinomas: differential diagnosis, prognostic factors and outcome report in 27 cases classified as classic and mixed variants. Appl Cancer Res. 2017;37:3.

    Google ScholarĀ 

  413. Cubilla AL, Velazquez EF, Young RH. Pseudohyperplastic squamous cell carcinoma of the penis associated with lichen sclerosus. An extremely well-differentiated, nonverruciform neoplasm that preferentially affects the foreskin and is frequently misdiagnosed: a report of 10 cases of a distinctive clinicopathologic entity. Am J Surg Pathol. 2004;28:895ā€“900.

    PubMedĀ  Google ScholarĀ 

  414. Cubilla AL, Ayala MT, Barreto JE, et al. Surface adenosquamous carcinoma of the penis. A report of three cases. Am J Surg Pathol. 1996;20:156ā€“60.

    PubMedĀ  CASĀ  Google ScholarĀ 

  415. Jamieson NV, Bullock KN, Barker TH. Adenosquamous carcinoma of the penis associated with balanitis xerotica obliterans. Br J Urol. 1986;58:730ā€“1.

    PubMedĀ  CASĀ  Google ScholarĀ 

  416. Masera A, Ovcak Z, Volavsek M, et al. Adenosquamous carcinoma of the penis. J Urol. 1997;157:2261.

    PubMedĀ  CASĀ  Google ScholarĀ 

  417. Velazquez EF, Melamed J, Barreto JE, et al. Sarcomatoid carcinoma of the penis: a clinicopathologic study of 15 cases. Am J Surg Pathol. 2005;29:1152ā€“8.

    PubMedĀ  Google ScholarĀ 

  418. Lont AP, Gallee MP, Snijders P, et al. Sarcomatoid squamous cell carcinoma of the penis: a clinical and pathological study of 5 cases. J Urol. 2004;172:932ā€“5.

    PubMedĀ  CASĀ  Google ScholarĀ 

  419. Cubilla AL, Reuter VE, Gregoire L, et al. Basaloid squamous cell carcinoma: a distinctive human papilloma virus-related penile neoplasm: a report of 20 cases. Am J Surg Pathol. 1998;22:755ā€“61.

    PubMedĀ  CASĀ  Google ScholarĀ 

  420. Mhawech P, Uchida T, Pelte MF. Immunohistochemical profile of high-grade urothelial bladder carcinoma and prostate adenocarcinoma. Hum Pathol. 2002;33:1136ā€“40.

    PubMedĀ  Google ScholarĀ 

  421. Oh WJ, Chung AM, Kim JS, et al. Differential immunohistochemical profiles for distinguishing prostate carcinoma and urothelial carcinoma. J Pathol Transl Med. 2016;50:345ā€“54.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  422. Cubilla AL, Lloveras B, Alemany L, et al. Basaloid squamous cell carcinoma of the penis with papillary features: a clinicopathologic study of 12 cases. Am J Surg Pathol. 2012;36:869ā€“75.

    PubMedĀ  Google ScholarĀ 

  423. Chaux A, Tamboli P, Ayala A, et al. Warty-basaloid carcinoma: clinicopathological features of a distinctive penile neoplasm. Report of 45 cases. Mod Pathol. 2010;23:896ā€“904.

    PubMedĀ  Google ScholarĀ 

  424. Liegl B, Regauer S. Penile clear cell carcinoma: a report of 5 cases of a distinct entity. Am J Surg Pathol. 2004;28:1513ā€“7.

    PubMedĀ  Google ScholarĀ 

  425. Sanchez DF, Rodriguez IM, Piris A, et al. Clear cell carcinoma of the penis: an HPV-related variant of squamous cell carcinoma: a report of 3 cases. Am J Surg Pathol. 2016;40:917ā€“22.

    PubMedĀ  Google ScholarĀ 

  426. Chaux A, Amin M, Cubilla AL, et al. Metastatic tumors to the penis: a report of 17 cases and review of the literature. Int J Surg Pathol. 2011;19:597ā€“606.

    PubMedĀ  Google ScholarĀ 

  427. Mentrikoski MJ, Frierson HF Jr, Stelow EB, et al. Lymphoepithelioma-like carcinoma of the penis: association with human papilloma virus infection. Histopathology. 2014;64:312ā€“5.

    PubMedĀ  Google ScholarĀ 

  428. Katona TM, Lopez-Beltran A, MacLennan GT, et al. Soft tissue tumors of the penis: a review. Anal Quant Cytol Histol. 2006;28:193ā€“206.

    PubMedĀ  Google ScholarĀ 

  429. Dehner LP, Smith BH. Soft tissue tumors of the penis. A clinicopathologic study of 46 cases. Cancer. 1970;25:1431ā€“47.

    PubMedĀ  CASĀ  Google ScholarĀ 

  430. McKenney JK, Collins MH, Carretero AP, et al. Penile myointimoma in children and adolescents: a clinicopathologic study of 5 cases supporting a distinct entity. Am J Surg Pathol. 2007;31:1622ā€“6.

    PubMedĀ  Google ScholarĀ 

  431. Belis JA, Post GJ, Rochman SC, et al. Genitourinary leiomyomas. Urology. 1979;13:424ā€“9.

    PubMedĀ  CASĀ  Google ScholarĀ 

  432. Laskin WB, Fetsch JF, Davis CJ Jr, et al. Granular cell tumor of the penis: clinicopathologic evaluation of 9 cases. Hum Pathol. 2005;36:291ā€“8.

    PubMedĀ  Google ScholarĀ 

  433. Fetsch JF, Sesterhenn IA, Miettinen M, et al. Epithelioid hemangioma of the penis: a clinicopathologic and immunohistochemical analysis of 19 cases, with special reference to exuberant examples often confused with epithelioid hemangioendothelioma and epithelioid angiosarcoma. Am J Surg Pathol. 2004;28:523ā€“33.

    PubMedĀ  Google ScholarĀ 

  434. Ismail M, Damato S, Freeman A, et al. Epithelioid hemangioma of the penis: case report and review of literature. J Med Case Rep. 2011;5:260.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  435. Fetsch JF, Brinsko RW, Davis CJ Jr, et al. A distinctive myointimal proliferation ('myointimoma') involving the corpus spongiosum of the glans penis: a clinicopathologic and immunohistochemical analysis of 10 cases. Am J Surg Pathol. 2000;24:1524ā€“30.

    PubMedĀ  CASĀ  Google ScholarĀ 

  436. Fetsch JF, Weiss SW. Observations concerning the pathogenesis of epithelioid hemangioma (angiolymphoid hyperplasia). Mod Pathol. 1991;4:449ā€“55.

    PubMedĀ  CASĀ  Google ScholarĀ 

  437. Bryant J. Granular cell tumor of penis and scrotum. Urology. 1995;45:332ā€“4.

    PubMedĀ  CASĀ  Google ScholarĀ 

  438. Mentzel T, Calonje E, Wadden C, et al. Myxofibrosarcoma. Clinicopathologic analysis of 75 cases with emphasis on the low-grade variant. Am J Surg Pathol. 1996;20:391ā€“405.

    PubMedĀ  CASĀ  Google ScholarĀ 

  439. Akino T, Shinohara N, Hatanaka K, et al. Successful penile reconstruction after multimodal therapy in patients with primitive neuroectodermal tumor originating from the penis. Int J Urol. 2014;21:619ā€“21.

    PubMedĀ  Google ScholarĀ 

  440. Al-Rikabi AC, Diab AR, Buckai A, et al. Primary synovial sarcoma of the penis--case report and literature review. Scand J Urol Nephrol. 1999;33:413ā€“5.

    PubMedĀ  CASĀ  Google ScholarĀ 

  441. Shringarpure SS, Kumar A, Shankar S, et al. A case of primary extraosseous penile osteosarcoma and review of the literature. Indian J Urol. 2012;28:102ā€“4.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  442. Woldrich JM, Silberstein JL, Saltzstein SL, et al. Penile Kaposis sarcoma in the state of California. Can J Urol. 2012;19:6178ā€“82.

    PubMedĀ  Google ScholarĀ 

  443. Fetsch JF, Davis CJ Jr, Miettinen M, et al. Leiomyosarcoma of the penis: a clinicopathologic study of 14 cases with review of the literature and discussion of the differential diagnosis. Am J Surg Pathol. 2004;28:115ā€“25.

    PubMedĀ  Google ScholarĀ 

  444. Buechner SA. Common skin disorders of the penis. BJU Int. 2002;90:498ā€“506; 450.

    Google ScholarĀ 

  445. Singh S, Bunker C. Male genital dermatoses in old age. Age Ageing 2008;37:500ā€“4.

    Google ScholarĀ 

  446. Giuliano AR, Tortolero-Luna G, Ferrer E, et al. Epidemiology of human papillomavirus infection in men, cancers other than cervical and benign conditions. Vaccine. 2008;26(Suppl 10):K17ā€“28.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  447. FernĆ”ndez-Nestosa MJ, GuimerĆ  N, Sanchez DF, et al. Human Papillomavirus (HPV) genotypes in condylomas, intraepithelial neoplasia, and invasive carcinoma of the penis using laser capture microdissection (LCM)-PCR: a study of 191 lesions in 43 patients. Am J Surg Pathol. 2017;41(6):820ā€“32.

    PubMedĀ  Google ScholarĀ 

  448. Schmauz R, Owor R. Condylomatous tumours of vulva, vagina, and penis. Relation between histological appearance and age. J Clin Pathol. 1980;33:1039ā€“46.

    PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  449. Schmauz R, Owor R. Epidemiology of malignant degeneration of condylomata acuminata in Uganda. Pathol Res Pract. 1980;170:91ā€“103.

    PubMedĀ  CASĀ  Google ScholarĀ 

  450. Bennetts LE, Wagner M, Giuliano AR, et al. Associations of anogenital low-risk human papillomavirus infection with cancer and acquisition of HIV. Sex Transm Dis. 2015;42:541ā€“4.

    PubMedĀ  CASĀ  Google ScholarĀ 

  451. Guimera N, Lloveras B, Lindeman J, et al. The occasional role of low-risk human papillomaviruses 6, 11, 42, 44, and 70 in anogenital carcinoma defined by laser capture microdissection/PCR methodology: results from a global study. Am J Surg Pathol. 2013;37:1299ā€“310.

    PubMedĀ  Google ScholarĀ 

  452. Loning T, Riviere A, Henke RP, et al. Penile/anal condylomas and squamous cell cancer. A HPV DNA hybridization study. Virchows Arch A Pathol Anat Histopathol. 1988;413:491ā€“8.

    PubMedĀ  CASĀ  Google ScholarĀ 

  453. del Pino M, Bleeker MC, Quint WG, et al. Comprehensive analysis of human papillomavirus prevalence and the potential role of low-risk types in verrucous carcinoma. Mod Pathol. 2012;25:1354ā€“63.

    PubMedĀ  Google ScholarĀ 

  454. Schmauz R, Findlay M, Lalwak A, et al. Variation in the appearance of giant condyloma in an Ungandan series of cases of carcinoma of the penis. Cancer. 1977;40:1686ā€“96.

    PubMedĀ  CASĀ  Google ScholarĀ 

  455. Suwa M, Takeda M, Bilim V, et al. Epidermoid cyst of the penis: a case report and review of the literature. Int J Urol. 2000;7:431ā€“3.

    PubMedĀ  CASĀ  Google ScholarĀ 

  456. Park HJ, Park NC, Park SW, et al. Penile epidermal inclusion cyst: a late complication of penile girth enhancement surgery. J Sex Med. 2008;5:2238ā€“40.

    PubMedĀ  Google ScholarĀ 

  457. Rattan J, Rattan S, Gupta DK. Epidermoid cyst of the penis with extension into the pelvis. J Urol. 1997;158:593.

    PubMedĀ  CASĀ  Google ScholarĀ 

  458. Faridi MM, Adhami S. Prepucial Epstein pearls. Indian J Pediatr. 1989;56:653ā€“5.

    PubMedĀ  CASĀ  Google ScholarĀ 

  459. Shiraki IW. Parametal cysts of the glans penis: a report of 9 cases. J Urol. 1975;114:544ā€“8.

    PubMedĀ  CASĀ  Google ScholarĀ 

  460. Lezcano C, Chaux A, Velazquez EF, et al. Clinicopathological features and histogenesis of penile cysts. Semin Diagn Pathol. 2015;32:245ā€“8.

    PubMedĀ  Google ScholarĀ 

  461. Hayashi T, Tsuda N, Shimada O, et al. A clinicopathologic study of tumors and tumor-like lesions of the penis. Acta Pathol Jpn. 1990;40:343ā€“51.

    PubMedĀ  CASĀ  Google ScholarĀ 

  462. Garcia Serrado D, Dominguez Hinarejos C, Martinez Verdu M, et al. Parameatal urethral cysts. 3 new paediatric cases and literature review. Actas Urol Esp. 2007;31:61ā€“3.

    PubMedĀ  CASĀ  Google ScholarĀ 

  463. Suarez Cuenca JA, Sanchez Garza M, Calderon Ferro F, et al. Parameatal cyst: presentation of two cases and a review of the literature. Arch Esp Urol. 1998;51:716ā€“8.

    PubMedĀ  CASĀ  Google ScholarĀ 

  464. Giannakopoulos S, Makris NE, Kalaitzis C, et al. Two unusual cases of median raphe penile cysts. Eur J Dermatol. 2007;17:342ā€“3.

    PubMedĀ  Google ScholarĀ 

  465. Nagore E, Sanchez-Motilla JM, Febrer MI, et al. Median raphe cysts of the penis: a report of five cases. Pediatr Dermatol. 1998;15:191ā€“3.

    PubMedĀ  CASĀ  Google ScholarĀ 

  466. Barua JM, Bates AW, Baithun SI. Squamous cell carcinoma arising in a para-urethral cyst. Br J Urol. 1997;80:158ā€“9.

    PubMedĀ  CASĀ  Google ScholarĀ 

  467. Tomasini C, Aloi F, Puiatti P, et al. Dermoid cyst of the penis. Dermatology. 1997;194:188ā€“90.

    PubMedĀ  CASĀ  Google ScholarĀ 

  468. Colonna M, Amadeo G, Ferlazzo G, et al. Multicystic lymphatic malformations of the penis. Report of two cases. Scand J Plast Reconstr Surg Hand Surg. 1992;26:235ā€“8.

    PubMedĀ  CASĀ  Google ScholarĀ 

  469. Beutler BD, Cohen PR. Verruciform Genital-Associated (Vegas) Xanthoma: report of a patient with verruciform xanthoma of the scrotum and literature review. Dermatol Online J. 2015;15:21(8).

    Google ScholarĀ 

  470. Stiff KM, Cohen PR. Vegas (Verruciform Genital-Associated) Xanthoma: a comprehensive literature review. Dermatol Ther (Heidelb). 2017;7(1):65ā€“79.

    Google ScholarĀ 

  471. Miake S, Nakahara T, Kurihara Y, et al. Verruciform xanthoma of the glans penis mimicking squamous cell carcinoma - role of scavenger receptor positive macrophages. Eur J Dermatol. 2012;22:391ā€“2.

    PubMedĀ  Google ScholarĀ 

  472. Mohsin SK, Lee MW, Amin MB, et al. Cutaneous verruciform xanthoma: a report of five cases investigating the etiology and nature of xanthomatous cells. Am J Surg Pathol. 1998;22:479ā€“87.

    PubMedĀ  CASĀ  Google ScholarĀ 

  473. Brock G, Hsu GL, Nunes L, et al. The anatomy of the tunica albuginea in the normal penis and Peyronie's disease. J Urol. 1997;157:276ā€“81.

    PubMedĀ  CASĀ  Google ScholarĀ 

  474. Al-Thakafi S, Al-Hathal N. Peyronieā€™s disease: a literature review on epidemiology, genetics, pathophysiology, diagnosis and work-up. Transl Androl Urol. 2016;5:280ā€“9.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  475. Stuntz M, Perlaky A, des Vignes F, et al. The prevalence of peyronieā€™s disease in the United States: a population-based study. PLoS One. 2016;11:e0150157.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  476. Davis CJ Jr. The microscopic pathology of Peyronieā€™s disease. J Urol. 1997;157:282ā€“4.

    PubMedĀ  Google ScholarĀ 

  477. Rane V, Read T. Penile appearance, lumps and bumps. Aust Fam Physician. 2013;42:270ā€“4.

    PubMedĀ  Google ScholarĀ 

  478. Ozeki M, Saito R, Tanaka M. Dermoscopic features of pearly penile papules. Dermatology. 2008;217:21ā€“2.

    PubMedĀ  Google ScholarĀ 

  479. Oertel YC, Johnson FB. Sclerosing lipogranuloma of male genitalia. Review of 23 cases. Arch Pathol Lab Med. 1977;101:321ā€“6.

    PubMedĀ  CASĀ  Google ScholarĀ 

  480. Foxton G, Vinciullo C, Tait CP, et al. Sclerosing lipogranuloma of the penis. Australas J Dermatol. 2011;52:e12ā€“4.

    PubMedĀ  Google ScholarĀ 

  481. Kantere D, Alvergren G, Gillstedt M, et al. Clinical features, complications and autoimmunity in male lichen sclerosus. Acta Derm Venereol. 2017;97(3):365ā€“69.

    Google ScholarĀ 

  482. Piris A, Sanchez DF, Canete S, et al. Pathological features of penile lichen sclerosus (LS) according to a topographical method of evaluation in 200 patients. Mod Pathol. 2015;28:251A.

    Google ScholarĀ 

  483. Barbagli G, Palminteri E, Mirri F, et al. Penile carcinoma in patients with genital lichen sclerosus: a multicenter survey. J Urol. 2006;175:1359ā€“63.

    PubMedĀ  CASĀ  Google ScholarĀ 

  484. Powell J, Robson A, Cranston D, et al. High incidence of lichen sclerosus in patients with squamous cell carcinoma of the penis. Br J Dermatol. 2001;145:85ā€“9.

    PubMedĀ  CASĀ  Google ScholarĀ 

  485. Nasca MR, Innocenzi D, Micali G. Penile cancer among patients with genital lichen sclerosus. J Am Acad Dermatol. 1999;41:911ā€“4.

    PubMedĀ  CASĀ  Google ScholarĀ 

  486. Pompeo A, Molina WR, Pohlman GD, Sehrt D, Kim FJ. Idiopathic scrotal calcinosis: a rare entity and a review of the literature. Can Urol Assoc J. 2013;7(5ā€“6):E439ā€“41.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  487. Yahya H, Rafindadi AH. Idiopathic scrotal calcinosis: a report of four cases and review of the literature. Int J Dermatol. 2005;44(3):206ā€“9.

    PubMedĀ  Google ScholarĀ 

  488. Lowe FC. Squamous cell carcinoma of the scrotum. J Urol. 1983;130(3):423ā€“7.

    PubMedĀ  CASĀ  Google ScholarĀ 

  489. Matoso A, Fabre V, Quddus MR, Lepe M, Lombardo KA, Manna P, et al. Prevalence and distribution of 15 high-risk human papillomavirus types in squamous cell carcinoma of the scrotum. Hum Pathol. 2016;53:130ā€“6.

    PubMedĀ  Google ScholarĀ 

  490. Vyas R, Zargar H, Trolio RD, Di Lorenzo G, Autorino R. Squamous cell carcinoma of the scrotum: a look beyond the chimneystacks. World J Clin Cases. 2014;2(11):654ā€“60.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  491. Dai B, Kong YY, Ye DW, Xu XW, Yao XD, Zhang SL. Basal cell carcinoma of the scrotum: clinicopathologic analysis of 10 cases. Dermatol Surg. 2012;38(5):783ā€“90.

    PubMedĀ  CASĀ  Google ScholarĀ 

  492. Gibson GE, Ahmed I. Perianal and genital basal cell carcinoma: a clinicopathologic review of 51 cases. J Am Acad Dermatol. 2001;45(1):68ā€“71.

    PubMedĀ  CASĀ  Google ScholarĀ 

  493. Shu B, Shen XX, Chen P, Fang XZ, Guo YL, Kong YY. Primary invasive extramammary Paget disease on penoscrotum: a clinicopathological analysis of 41 cases. Hum Pathol. 2016;47(1):70ā€“7.

    PubMedĀ  Google ScholarĀ 

  494. Williamson JD, Colome MI, Sahin A, Ayala AG, Medeiros LJ. Pagetoid bowen disease: a report of 2 cases that express cytokeratin 7. Arch Pathol Lab Med. 2000;124(3):427ā€“30.

    PubMedĀ  CASĀ  Google ScholarĀ 

  495. Nowak MA, Guerriere-Kovach P, Pathan A, Campbell TE, Deppisch LM. Perianal Pagetā€™s disease: distinguishing primary and secondary lesions using immunohistochemical studies including gross cystic disease fluid protein-15 and cytokeratin 20 expression. Arch Pathol Lab Med. 1998;122(12):1077ā€“81.

    PubMedĀ  CASĀ  Google ScholarĀ 

  496. Battles OE, Page DL, Johnson JE. Cytokeratins, CEA, and mucin histochemistry in the diagnosis and characterization of extramammary Pagetā€™s disease. Am J Clin Pathol. 1997;108(1):6ā€“12.

    PubMedĀ  CASĀ  Google ScholarĀ 

  497. Talamo TS, Shonnard JW. Small renal adenocarcinoma with metastases. J Urol. 1980;124(1):132ā€“4.

    PubMedĀ  CASĀ  Google ScholarĀ 

  498. Thoenes W, Storkel S, Rumpelt HJ. Histopathology and classification of renal cell tumors (adenomas, oncocytomas and carcinomas). The basic cytological and histopathological elements and their use for diagnostics. Pathol Res Pract. 1986;181(2):125ā€“43.

    PubMedĀ  CASĀ  Google ScholarĀ 

  499. Bostwick DG, Murphy GP. Diagnosis and prognosis of renal cell carcinoma: highlights from an international consensus workshop. Semin Urol Oncol. 1998;16(1):46ā€“52.

    PubMedĀ  CASĀ  Google ScholarĀ 

  500. Wang KL, Weinrach DM, Luan C, Han M, Lin F, Teh BT, et al. Renal papillary adenoma--a putative precursor of papillary renal cell carcinoma. Hum Pathol. 2007;38(2):239ā€“46.

    PubMedĀ  CASĀ  Google ScholarĀ 

  501. Amin MB, Crotty TB, Tickoo SK, Farrow GM. Renal oncocytoma: a reappraisal of morphologic features with clinicopathologic findings in 80 cases. Am J Surg Pathol. 1997;21(1):1ā€“12.

    PubMedĀ  CASĀ  Google ScholarĀ 

  502. Lieber MM, Tomera KM, Farrow GM. Renal oncocytoma. J Urol. 1981;125(4):481ā€“5.

    PubMedĀ  CASĀ  Google ScholarĀ 

  503. Trpkov K, Yilmaz A, Uzer D, Dishongh KM, Quick CM, Bismar TA, et al. Renal oncocytoma revisited: a clinicopathological study of 109 cases with emphasis on problematic diagnostic features. Histopathology. 2010;57(6):893ā€“906.

    PubMedĀ  Google ScholarĀ 

  504. Dechet CB, Bostwick DG, Blute ML, Bryant SC, Zincke H. Renal oncocytoma: multifocality, bilateralism, metachronous tumor development and coexistent renal cell carcinoma. J Urol. 1999;162(1):40ā€“2.

    PubMedĀ  CASĀ  Google ScholarĀ 

  505. Perez-Ordonez B, Hamed G, Campbell S, Erlandson RA, Russo P, Gaudin PB, et al. Renal oncocytoma: a clinicopathologic study of 70 cases. Am J Surg Pathol. 1997;21(8):871ā€“83.

    PubMedĀ  CASĀ  Google ScholarĀ 

  506. Yusenko MV. Molecular pathology of renal oncocytoma: a review. Int J Urol. 2010;17:602. 2011;18(8):614.

    Google ScholarĀ 

  507. Muir TE, Cheville JC, Lager DJ. Metanephric adenoma, nephrogenic rests, and Wilms' tumor: a histologic and immunophenotypic comparison. Am J Surg Pathol. 2001;25(10):1290ā€“6.

    PubMedĀ  CASĀ  Google ScholarĀ 

  508. Argani P. Metanephric neoplasms: the hyperdifferentiated, benign end of the Wilms tumor spectrum? Clin Lab Med. 2005;25(2):379ā€“92.

    PubMedĀ  Google ScholarĀ 

  509. Davis CJ Jr, Barton JH, Sesterhenn IA, Mostofi FK. Metanephric adenoma. Clinicopathological study of fifty patients. Am J Surg Pathol. 1995;19(10):1101ā€“14.

    PubMedĀ  Google ScholarĀ 

  510. Srigley JR, Delahunt B, Eble JN, Egevad L, Epstein JI, Grignon D, et al. The International Society of Urological Pathology (ISUP) Vancouver classification of renal neoplasia. Am J Surg Pathol. 2013;37(10):1469ā€“89.

    PubMedĀ  Google ScholarĀ 

  511. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016;66(1):7ā€“30.

    PubMedĀ  Google ScholarĀ 

  512. Brinker DA, Amin MB, de Peralta-Venturina M, Reuter V, Chan DY, Epstein JI. Extensively necrotic cystic renal cell carcinoma: a clinicopathologic study with comparison to other cystic and necrotic renal cancers. Am J Surg Pathol. 2000;24(7):988ā€“95.

    PubMedĀ  CASĀ  Google ScholarĀ 

  513. Truong LD, Shen SS. Immunohistochemical diagnosis of renal neoplasms. Arch Pathol Lab Med. 2011;135(1):92ā€“109.

    PubMedĀ  Google ScholarĀ 

  514. Skinnider BF, Folpe AL, Hennigar RA, Lim SD, Cohen C, Tamboli P, et al. Distribution of cytokeratins and vimentin in adult renal neoplasms and normal renal tissue: potential utility of a cytokeratin antibody panel in the differential diagnosis of renal tumors. Am J Surg Pathol. 2005;29(6):747ā€“54.

    PubMedĀ  Google ScholarĀ 

  515. Ross H, Martignoni G, Argani P. Renal cell carcinoma with clear cell and papillary features. Arch Pathol Lab Med. 2012;136(4):391ā€“9.

    PubMedĀ  Google ScholarĀ 

  516. Rigola MA, Casadevall C, Bernues M, Caballin MR, Fuster C, Gelabert A, et al. Analysis of kidney tumors by comparative genomic hybridization and conventional cytogenetics. Cancer Genet Cytogenet. 2002;137(1):49ā€“53.

    PubMedĀ  CASĀ  Google ScholarĀ 

  517. Furge KA, Lucas KA, Takahashi M, Sugimura J, Kort EJ, Kanayama HO, et al. Robust classification of renal cell carcinoma based on gene expression data and predicted cytogenetic profiles. Cancer Res. 2004;64(12):4117ā€“21.

    PubMedĀ  CASĀ  Google ScholarĀ 

  518. Shuch B, Bratslavsky G, Linehan WM, Srinivasan R. Sarcomatoid renal cell carcinoma: a comprehensive review of the biology and current treatment strategies. Oncologist. 2012;17(1):46ā€“54.

    PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  519. de Peralta-Venturina M, Moch H, Amin M, Tamboli P, Hailemariam S, Mihatsch M, et al. Sarcomatoid differentiation in renal cell carcinoma: a study of 101 cases. Am J Surg Pathol. 2001;25(3):275ā€“84.

    PubMedĀ  Google ScholarĀ 

  520. Shuch B, Said J, LaRochelle JC, Zhou Y, Li G, Klatte T, et al. Histologic evaluation of metastases in renal cell carcinoma with sarcomatoid transformation and its implications for systemic therapy. Cancer. 2010;116(3):616ā€“24.

    PubMedĀ  Google ScholarĀ 

  521. Halat S, Eble JN, Grignon DJ, Lopez-Beltran A, Montironi R, Tan PH, et al. Multilocular cystic renal cell carcinoma is a subtype of clear cell renal cell carcinoma. Mod Pathol. 2010;23(7):931ā€“6.

    PubMedĀ  Google ScholarĀ 

  522. Delahunt B, Eble JN. Papillary renal cell carcinoma: a clinicopathologic and immunohistochemical study of 105 tumors. Mod Pathol. 1997;10(6):537ā€“44.

    PubMedĀ  CASĀ  Google ScholarĀ 

  523. Udager AM, Mehra R. Morphologic, molecular, and taxonomic evolution of renal cell carcinoma: a conceptual perspective with emphasis on updates to the 2016 World Health Organization Classification. Arch Pathol Lab Med. 2016;140(10):1026ā€“37.

    PubMedĀ  CASĀ  Google ScholarĀ 

  524. Tickoo SK, de Peralta-Venturina MN, Harik LR, Worcester HD, Salama ME, Young AN, et al. Spectrum of epithelial neoplasms in end-stage renal disease: an experience from 66 tumor-bearing kidneys with emphasis on histologic patterns distinct from those in sporadic adult renal neoplasia. Am J Surg Pathol. 2006;30(2):141ā€“53.

    PubMedĀ  Google ScholarĀ 

  525. Zhou H, Zheng S, Truong LD, Ro JY, Ayala AG, Shen SS. Clear cell papillary renal cell carcinoma is the fourth most common histologic type of renal cell carcinoma in 290 consecutive nephrectomies for renal cell carcinoma. Hum Pathol. 2014;45(1):59ā€“64.

    PubMedĀ  Google ScholarĀ 

  526. Aydin H, Chen L, Cheng L, Vaziri S, He H, Ganapathi R, et al. Clear cell tubulopapillary renal cell carcinoma: a study of 36 distinctive low-grade epithelial tumors of the kidney. Am J Surg Pathol. 2010;34(11):1608ā€“21.

    PubMedĀ  Google ScholarĀ 

  527. Michal M, Hes O, Nemcova J, Sima R, Kuroda N, Bulimbasic S, et al. Renal angiomyoadenomatous tumor: morphologic, immunohistochemical, and molecular genetic study of a distinct entity. Virchows Arch. 2009;454(1):89ā€“99.

    PubMedĀ  CASĀ  Google ScholarĀ 

  528. Petersson F, Grossmann P, Hora M, Sperga M, Montiel DP, Martinek P, et al. Renal cell carcinoma with areas mimicking renal angiomyoadenomatous tumor/clear cell papillary renal cell carcinoma. Hum Pathol. 2013;44(7):1412ā€“20.

    PubMedĀ  Google ScholarĀ 

  529. Michal M, Hes O, Havlicek F. Benign renal angiomyoadenomatous tumor: a previously unreported renal tumor. Ann Diagn Pathol. 2000;4(5):311ā€“5.

    PubMedĀ  CASĀ  Google ScholarĀ 

  530. Williamson SR, Eble JN, Cheng L, Grignon DJ. Clear cell papillary renal cell carcinoma: differential diagnosis and extended immunohistochemical profile. Mod Pathol. 2013;26(5):697ā€“708.

    PubMedĀ  CASĀ  Google ScholarĀ 

  531. Rao P, Monzon F, Jonasch E, Matin SF, Tamboli P. Clear cell papillary renal cell carcinoma in patients with von Hippel-Lindau syndrome--clinicopathological features and comparative genomic analysis of 3 cases. Hum Pathol. 2014;45(9):1966ā€“72.

    PubMedĀ  CASĀ  Google ScholarĀ 

  532. Williamson SR, Zhang S, Eble JN, Grignon DJ, Martignoni G, Brunelli M, et al. Clear cell papillary renal cell carcinoma-like tumors in patients with von Hippel-Lindau disease are unrelated to sporadic clear cell papillary renal cell carcinoma. Am J Surg Pathol. 2013;37(8):1131ā€“9.

    PubMedĀ  Google ScholarĀ 

  533. Aron M, Chang E, Herrera L, Hes O, Hirsch MS, Comperat E, et al. Clear cell-papillary renal cell carcinoma of the kidney not associated with end-stage renal disease: clinicopathologic correlation with expanded immunophenotypic and molecular characterization of a large cohort with emphasis on relationship with renal angiomyoadenomatous tumor. Am J Surg Pathol. 2015;39(7):873ā€“88.

    PubMedĀ  Google ScholarĀ 

  534. Storkel S, Steart PV, Drenckhahn D, Thoenes W. The human chromophobe cell renal carcinoma: its probable relation to intercalated cells of the collecting duct. Virchows Arch B Cell Pathol Incl Mol Pathol. 1989;56(4):237ā€“45.

    PubMedĀ  CASĀ  Google ScholarĀ 

  535. Zbar B, Alvord WG, Glenn G, Turner M, Pavlovich CP, Schmidt L, et al. Risk of renal and colonic neoplasms and spontaneous pneumothorax in the Birt-Hogg-Dube syndrome. Cancer Epidemiol Biomark Prev. 2002;11(4):393ā€“400.

    Google ScholarĀ 

  536. Crotty TB, Farrow GM, Lieber MM. Chromophobe cell renal carcinoma: clinicopathological features of 50 cases. J Urol. 1995;154(3):964ā€“7.

    PubMedĀ  CASĀ  Google ScholarĀ 

  537. Hes O, Petersson F, Kuroda N, Hora M, Michal M. Renal hybrid oncocytic/chromophobe tumorsĀ ā€“ a review. Histol Histopathol. 2013;28(10):1257ā€“64.

    PubMedĀ  Google ScholarĀ 

  538. Pavlovich CP, Walther MM, Eyler RA, Hewitt SM, Zbar B, Linehan WM, et al. Renal tumors in the Birt-Hogg-Dube syndrome. Am J Surg Pathol. 2002;26(12):1542ā€“52.

    PubMedĀ  Google ScholarĀ 

  539. Pavlovich CP, Grubb RL 3rd, Hurley K, Glenn GM, Toro J, Schmidt LS, et al. Evaluation and management of renal tumors in the Birt-Hogg-Dube syndrome. J Urol. 2005;173(5):1482ā€“6.

    PubMedĀ  Google ScholarĀ 

  540. Reuter VE, Argani P, Zhou M, Delahunt B, Members of the IIiDUPG. Best practices recommendations in the application of immunohistochemistry in the kidney tumors: report from the International Society of Urologic Pathology consensus conference. Am J Surg Pathol. 2014;38(8):e35ā€“49.

    PubMedĀ  Google ScholarĀ 

  541. Davis CF, Ricketts CJ, Wang M, Yang L, Cherniack AD, Shen H, et al. The somatic genomic landscape of chromophobe renal cell carcinoma. Cancer Cell. 2014;26(3):319ā€“30.

    PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  542. Durinck S, Stawiski EW, Pavia-Jimenez A, Modrusan Z, Kapur P, Jaiswal BS, et al. Spectrum of diverse genomic alterations define non-clear cell renal carcinoma subtypes. Nat Genet. 2015;47(1):13ā€“21.

    PubMedĀ  CASĀ  Google ScholarĀ 

  543. Kato I, Iribe Y, Nagashima Y, Kuroda N, Tanaka R, Nakatani Y, et al. Fluorescent and chromogenic in situ hybridization of CEN17q as a potent useful diagnostic marker for Birt-Hogg-Dube syndrome-associated chromophobe renal cell carcinomas. Hum Pathol. 2016;52:74ā€“82.

    PubMedĀ  CASĀ  Google ScholarĀ 

  544. Fleming S, Lewi HJ. Collecting duct carcinoma of the kidney. Histopathology. 1986;10(11):1131ā€“41.

    PubMedĀ  CASĀ  Google ScholarĀ 

  545. Albadine R, Schultz L, Illei P, Ertoy D, Hicks J, Sharma R, et al. PAX8 (+)/p63 (āˆ’) immunostaining pattern in renal collecting duct carcinoma (CDC): a useful immunoprofile in the differential diagnosis of CDC versus urothelial carcinoma of upper urinary tract. Am J Surg Pathol. 2010;34(7):965ā€“9.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  546. Gonzalez-Roibon N, Albadine R, Sharma R, Faraj SF, Illei PB, Argani P, et al. The role of GATA binding protein 3 in the differential diagnosis of collecting duct and upper tract urothelial carcinomas. Hum Pathol. 2013;44(12):2651ā€“7.

    PubMedĀ  CASĀ  Google ScholarĀ 

  547. Seo AN, Yoon G, Ro JY. Clinicopathologic and molecular pathology of collecting duct carcinoma and related renal cell carcinomas. Adv Anat Pathol. 2017;24(2):65ā€“77.

    PubMedĀ  CASĀ  Google ScholarĀ 

  548. Calderaro J, Moroch J, Pierron G, Pedeutour F, Grison C, Maille P, et al. SMARCB1/INI1 inactivation in renal medullary carcinoma. Histopathology. 2012;61(3):428ā€“35.

    PubMedĀ  Google ScholarĀ 

  549. Rao P, Tannir NM, Tamboli P. Expression of OCT3/4 in renal medullary carcinoma represents a potential diagnostic pitfall. Am J Surg Pathol. 2012;36(4):583ā€“8.

    PubMedĀ  Google ScholarĀ 

  550. Argani P. MiT family translocation renal cell carcinoma. Semin Diagn Pathol. 2015;32(2):103ā€“13.

    PubMedĀ  Google ScholarĀ 

  551. Komai Y, Fujiwara M, Fujii Y, Mukai H, Yonese J, Kawakami S, et al. Adult Xp11 translocation renal cell carcinoma diagnosed by cytogenetics and immunohistochemistry. Clin Cancer Res. 2009;15(4):1170ā€“6.

    PubMedĀ  CASĀ  Google ScholarĀ 

  552. Zhong M, De Angelo P, Osborne L, Keane-Tarchichi M, Goldfischer M, Edelmann L, et al. Dual-color, break-apart FISH assay on paraffin-embedded tissues as an adjunct to diagnosis of Xp11 translocation renal cell carcinoma and alveolar soft part sarcoma. Am J Surg Pathol. 2010;34(6):757ā€“66.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  553. Argani P, Lae M, Hutchinson B, Reuter VE, Collins MH, Perentesis J, et al. Renal carcinomas with the t(6;11)(p21;q12): clinicopathologic features and demonstration of the specific alpha-TFEB gene fusion by immunohistochemistry, RT-PCR, and DNA PCR. Am J Surg Pathol. 2005;29(2):230ā€“40.

    PubMedĀ  Google ScholarĀ 

  554. Sukov WR, Hodge JC, Lohse CM, Leibovich BC, Thompson RH, Pearce KE, et al. TFE3 rearrangements in adult renal cell carcinoma: clinical and pathologic features with outcome in a large series of consecutively treated patients. Am J Surg Pathol. 2012;36(5):663ā€“70.

    PubMedĀ  Google ScholarĀ 

  555. Lopez-Beltran A, Scarpelli M, Montironi R, Kirkali Z. 2004 WHO classification of the renal tumors of the adults. Eur Urol. 2006;49(5):798ā€“805.

    PubMedĀ  Google ScholarĀ 

  556. Fine SW, Argani P, DeMarzo AM, Delahunt B, Sebo TJ, Reuter VE, et al. Expanding the histologic spectrum of mucinous tubular and spindle cell carcinoma of the kidney. Am J Surg Pathol. 2006;30(12):1554ā€“60.

    PubMedĀ  Google ScholarĀ 

  557. Dhillon J, Amin MB, Selbs E, Turi GK, Paner GP, Reuter VE. Mucinous tubular and spindle cell carcinoma of the kidney with sarcomatoid change. Am J Surg Pathol. 2009;33(1):44ā€“9.

    PubMedĀ  Google ScholarĀ 

  558. Asghar AM, Uhlman MA, Dahmoush L, Deorah S. Osseous metaplasia in mucinous tubular and spindle cell carcinoma of the kidney: a case of massive, bilateral tumors. Case Rep Urol. 2015;2015:465450.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  559. Mehra R, Vats P, Cieslik M, Cao X, Su F, Shukla S, et al. Biallelic alteration and dysregulation of the hippo pathway in mucinous tubular and spindle cell carcinoma of the kidney. Cancer Discov. 2016;6(11):1258ā€“66.

    PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  560. Amin MB, MacLennan GT, Gupta R, Grignon D, Paraf F, Vieillefond A, et al. Tubulocystic carcinoma of the kidney: clinicopathologic analysis of 31 cases of a distinctive rare subtype of renal cell carcinoma. Am J Surg Pathol. 2009;33(3):384ā€“92.

    PubMedĀ  Google ScholarĀ 

  561. Yang XJ, Zhou M, Hes O, Shen S, Li R, Lopez J, et al. Tubulocystic carcinoma of the kidney: clinicopathologic and molecular characterization. Am J Surg Pathol. 2008;32(2):177ā€“87.

    PubMedĀ  Google ScholarĀ 

  562. Launonen V, Vierimaa O, Kiuru M, Isola J, Roth S, Pukkala E, et al. Inherited susceptibility to uterine leiomyomas and renal cell cancer. Proc Natl Acad Sci U S A. 2001;98(6):3387ā€“92.

    PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  563. Merino MJ, Torres-Cabala C, Pinto P, Linehan WM. The morphologic spectrum of kidney tumors in hereditary leiomyomatosis and renal cell carcinoma (HLRCC) syndrome. Am J Surg Pathol. 2007;31(10):1578ā€“85.

    PubMedĀ  Google ScholarĀ 

  564. Chen YB, Brannon AR, Toubaji A, Dudas ME, Won HH, Al-Ahmadie HA, et al. Hereditary leiomyomatosis and renal cell carcinoma syndrome-associated renal cancer: recognition of the syndrome by pathologic features and the utility of detecting aberrant succination by immunohistochemistry. Am J Surg Pathol. 2014;38(5):627ā€“37.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  565. Trpkov K, Hes O, Agaimy A, Bonert M, Martinek P, Magi-Galluzzi C, et al. Fumarate hydratase-deficient renal cell carcinoma is strongly correlated with fumarate hydratase mutation and hereditary leiomyomatosis and renal cell carcinoma syndrome. Am J Surg Pathol. 2016;40(7):865ā€“75.

    PubMedĀ  Google ScholarĀ 

  566. Foshat M, Eyzaguirre E. Acquired cystic disease-associated renal cell carcinoma: review of pathogenesis, morphology, ancillary tests, and clinical features. Arch Pathol Lab Med. 2017;141(4):600ā€“6.

    PubMedĀ  CASĀ  Google ScholarĀ 

  567. Amin MB, Gupta R, Ondrej H, McKenney JK, Michal M, Young AN, et al. Primary thyroid-like follicular carcinoma of the kidney: report of 6 cases of a histologically distinctive adult renal epithelial neoplasm. Am J Surg Pathol. 2009;33(3):393ā€“400.

    PubMedĀ  Google ScholarĀ 

  568. Dhillon J, Tannir NM, Matin SF, Tamboli P, Czerniak BA, Guo CC. Thyroid-like follicular carcinoma of the kidney with metastases to the lungs and retroperitoneal lymph nodes. Hum Pathol. 2011;42(1):146ā€“50.

    PubMedĀ  CASĀ  Google ScholarĀ 

  569. Gill AJ, Pachter NS, Chou A, Young B, Clarkson A, Tucker KM, et al. Renal tumors associated with germline SDHB mutation show distinctive morphology. Am J Surg Pathol. 2011;35(10):1578ā€“85.

    PubMedĀ  Google ScholarĀ 

  570. Gill AJ, Hes O, Papathomas T, Sedivcova M, Tan PH, Agaimy A, et al. Succinate dehydrogenase (SDH)-deficient renal carcinoma: a morphologically distinct entity: a clinicopathologic series of 36 tumors from 27 patients. Am J Surg Pathol. 2014;38(12):1588ā€“602.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  571. Mehra R, Smith SC, Divatia M, Amin MB. Emerging entities in renal neoplasia. Surg Pathol Clin. 2015;8(4):623ā€“56.

    PubMedĀ  Google ScholarĀ 

  572. Shen SS, Truong LD, Scarpelli M, Lopez-Beltran A. Role of immunohistochemistry in diagnosing renal neoplasms: when is it really useful? Arch Pathol Lab Med. 2012;136(4):410ā€“7.

    PubMedĀ  Google ScholarĀ 

  573. Fuhrman SA, Lasky LC, Limas C. Prognostic significance of morphologic parameters in renal cell carcinoma. Am J Surg Pathol. 1982;6(7):655ā€“63.

    PubMedĀ  CASĀ  Google ScholarĀ 

  574. Lohse CM, Blute ML, Zincke H, Weaver AL, Cheville JC. Comparison of standardized and nonstandardized nuclear grade of renal cell carcinoma to predict outcome among 2,042 patients. Am J Clin Pathol. 2002;118(6):877ā€“86.

    PubMedĀ  Google ScholarĀ 

  575. Turbiner J, Amin MB, Humphrey PA, Srigley JR, De Leval L, Radhakrishnan A, et al. Cystic nephroma and mixed epithelial and stromal tumor of kidney: a detailed clinicopathologic analysis of 34 cases and proposal for renal epithelial and stromal tumor (REST) as a unifying term. Am J Surg Pathol. 2007;31(4):489ā€“500.

    PubMedĀ  Google ScholarĀ 

  576. Xie WL, Lian JY, Li B, Tian XY, Li Z. Mixed epithelial and stromal tumor of kidney with renal vein extension: an unusual case report and review of literature. Histol Histopathol. 2017;32(4):361ā€“9.

    PubMedĀ  Google ScholarĀ 

  577. Michal M, Hes O, Bisceglia M, Simpson RH, Spagnolo DV, Parma A, et al. Mixed epithelial and stromal tumors of the kidney. A report of 22 cases. Virchows Arch. 2004;445(4):359ā€“67.

    PubMedĀ  Google ScholarĀ 

  578. Mudaliar KM, Mehta V, Gupta GN, Picken MM. Expanding the morphologic spectrum of adult biphasic renal tumors--mixed epithelial and stromal tumor of the kidney with focal papillary renal cell carcinoma: case report and review of the literature. Int J Surg Pathol. 2014;22(3):266ā€“71.

    PubMedĀ  Google ScholarĀ 

  579. Guo J, Tretiakova MS, Troxell ML, Osunkoya AO, Fadare O, Sangoi AR, et al. Tuberous sclerosis-associated renal cell carcinoma: a clinicopathologic study of 57 separate carcinomas in 18 patients. Am J Surg Pathol. 2014;38(11):1457ā€“67.

    PubMedĀ  Google ScholarĀ 

  580. L'Hostis H, Deminiere C, Ferriere JM, Coindre JM. Renal angiomyolipoma: a clinicopathologic, immunohistochemical, and follow-up study of 46 cases. Am J Surg Pathol. 1999;23(9):1011ā€“20.

    PubMedĀ  CASĀ  Google ScholarĀ 

  581. Durand X, Renard-Penna R, Comperat E, Bitker MO, Richard F. Renal angiomyolipoma associated with inferior vena cava thrombus. Case Rep Med. 2009;2009:789078.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  582. Fine SW, Reuter VE, Epstein JI, Argani P. Angiomyolipoma with epithelial cysts (AMLEC): a distinct cystic variant of angiomyolipoma. Am J Surg Pathol. 2006;30(5):593ā€“9.

    PubMedĀ  Google ScholarĀ 

  583. LeRoy MA, Rao P. Angiomyolipoma with epithelial cysts. Arch Pathol Lab Med. 2016;140(6):594ā€“7.

    PubMedĀ  CASĀ  Google ScholarĀ 

  584. Nese N, Martignoni G, Fletcher CD, Gupta R, Pan CC, Kim H, et al. Pure epithelioid PEComas (so-called epithelioid angiomyolipoma) of the kidney: a clinicopathologic study of 41 cases: detailed assessment of morphology and risk stratification. Am J Surg Pathol. 2011;35(2):161ā€“76.

    PubMedĀ  Google ScholarĀ 

  585. Gupta S, Jimenez RE, Folpe AL, Cheville JC. Renal leiomyoma and leiomyosarcoma: a study of 57 cases. Am J Surg Pathol. 2016;40(11):1557ā€“63.

    PubMedĀ  Google ScholarĀ 

  586. Brown JG, Folpe AL, Rao P, Lazar AJ, Paner GP, Gupta R, et al. Primary vascular tumors and tumor-like lesions of the kidney: a clinicopathologic analysis of 25 cases. Am J Surg Pathol. 2010;34(7):942ā€“9.

    PubMedĀ  Google ScholarĀ 

  587. Schoolmeester JK, Cheville JC, Folpe AL. Synovial sarcoma of the kidney: a clinicopathologic, immunohistochemical, and molecular genetic study of 16 cases. Am J Surg Pathol. 2014;38(1):60ā€“5.

    PubMedĀ  Google ScholarĀ 

  588. Kuroda N, Gotoda H, Ohe C, Mikami S, Inoue K, Nagashima Y, et al. Review of juxtaglomerular cell tumor with focus on pathobiological aspect. Diagn Pathol. 2011;6:80.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  589. Kim HJ, Kim CH, Choi YJ, Ayala AG, Amirikachi M, Ro JY. Juxtaglomerular cell tumor of kidney with CD34 and CD117 immunoreactivity: report of 5 cases. Arch Pathol Lab Med. 2006;130(5):707ā€“11.

    PubMedĀ  Google ScholarĀ 

  590. Martin SA, Mynderse LA, Lager DJ, Cheville JC. Juxtaglomerular cell tumor: a clinicopathologic study of four cases and review of the literature. Am J Clin Pathol. 2001;116(6):854ā€“63.

    PubMedĀ  CASĀ  Google ScholarĀ 

  591. Calio A, Warfel KA, Eble JN. Renomedullary interstitial cell tumors: pathologic features and clinical correlations. Am J Surg Pathol. 2016;40(12):1693ā€“701.

    PubMedĀ  Google ScholarĀ 

  592. Murugan P, Rao P, Tamboli P, Czerniak B, Guo CC. Primary Ewing sarcoma/primitive neuroectodermal tumor of the kidney: a clinicopathologic study of 23 cases. Pathol Oncol Res. 2018;24(1):153ā€“9.

    PubMedĀ  CASĀ  Google ScholarĀ 

  593. Joseph TJ, Becker DI, Turton AF. Renal malignant fibrous histiocytoma. Urology. 1991;37(5):483ā€“9.

    PubMedĀ  CASĀ  Google ScholarĀ 

  594. Ptochos A, Karydas G, Iosifidis N, Tyrothoulakis E, Papazafiriou G, Kehagia-Koutoufari T. Primary renal malignant fibrous histiocytoma. A case report and review of the literature. Urol Int. 1999;63(4):261ā€“4.

    PubMedĀ  CASĀ  Google ScholarĀ 

  595. Tarjan M, Cserni G, Szabo Z. Malignant fibrous histiocytoma of the kidney. Scand J Urol Nephrol. 2001;35(6):518ā€“20.

    PubMedĀ  CASĀ  Google ScholarĀ 

  596. Eroglu M, Bakirtas H, Cimentepe E, Unsal A, Ataoglu O, Balbay MD. Malignant fibrous histiocytoma arising from the renal capsule. Urol Int. 2005;75(4):368ā€“70.

    PubMedĀ  Google ScholarĀ 

  597. Gabbert H, Wagner R, Becht E. Malignant fibrous histiocytoma of the renal capsule. J Cancer Res Clin Oncol. 1981;100(3):285ā€“93.

    PubMedĀ  CASĀ  Google ScholarĀ 

  598. Garg P, Dass BK, Singh S. Malignant fibrous histiocytoma of renal capsule. J Indian Med Assoc. 1995;93(11):430. 2.

    PubMedĀ  CASĀ  Google ScholarĀ 

  599. Kitajima K, Kaji Y, Morita M, Okuda Y, Sugimura K. Malignant fibrous histiocytoma arising from the renal capsule. Magn Reson Med Sci. 2003;2(4):199ā€“202.

    PubMedĀ  Google ScholarĀ 

  600. Schneider JA, Konig H, Hertel E. Malignant fibrous histiocytoma of the renal capsule. Br J Urol. 1992;69(4):435ā€“6.

    PubMedĀ  CASĀ  Google ScholarĀ 

  601. Aung PP, Killian K, Poropatich CO, Linehan WM, Merino MJ. Primary neuroendocrine tumors of the kidney: morphological and molecular alterations of an uncommon malignancy. Hum Pathol. 2013;44(5):873ā€“80.

    PubMedĀ  Google ScholarĀ 

  602. Kuroda N, Tanaka A, Ohe C, Mikami S, Nagashima Y, Inoue K, et al. Review of renal carcinoid tumor with focus on clinical and pathobiological aspects. Histol Histopathol. 2013;28(1):15ā€“21.

    PubMedĀ  CASĀ  Google ScholarĀ 

  603. Lane BR, Jour G, Zhou M. Renal neuroendocrine tumors. Indian J Urol. 2009;25(2):155ā€“60.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  604. Sangoi AR, Ohgami RS, Pai RK, Beck AH, McKenney JK, Pai RK. PAX8 expression reliably distinguishes pancreatic well-differentiated neuroendocrine tumors from ileal and pulmonary well-differentiated neuroendocrine tumors and pancreatic acinar cell carcinoma. Mod Pathol. 2011;24(3):412ā€“24.

    PubMedĀ  CASĀ  Google ScholarĀ 

  605. Sotelo-Avila C, Gonzalez-Crussi F, Fowler JW. Complete and incomplete forms of Beckwith-Wiedemann syndrome: their oncogenic potential. J Pediatr. 1980;96(1):47ā€“50.

    PubMedĀ  CASĀ  Google ScholarĀ 

  606. Heppe RK, Koyle MA, Beckwith JB. Nephrogenic rests in Wilms tumor patients with the Drash syndrome. J Urol. 1991;145(6):1225ā€“8.

    PubMedĀ  CASĀ  Google ScholarĀ 

  607. Beckwith JB. Wilms' tumor and other renal tumors of childhood: a selective review from the National Wilms' Tumor Study Pathology Center. Hum Pathol. 1983;14(6):481ā€“92.

    PubMedĀ  CASĀ  Google ScholarĀ 

  608. Grimes MM, Wolff M, Wolff JA, Jaretzki A 3rd, Blanc WA. Ganglion cells in metastatic Wilms' tumor. Review of a histogenetic controversy. Am J Surg Pathol. 1982;6(6):565ā€“71.

    PubMedĀ  CASĀ  Google ScholarĀ 

  609. Fernandes ET, Parham DM, Ribeiro RC, Douglass EC, Kumar AP, Wilimas J. Teratoid Wilms' tumor: the St Jude experience. J Pediatr Surg. 1988;23(12):1131ā€“4.

    PubMedĀ  CASĀ  Google ScholarĀ 

  610. Vujanic GM, Harms D, Sandstedt B, Weirich A, de Kraker J, Delemarre JF. New definitions of focal and diffuse anaplasia in Wilms tumor: the International Society of Paediatric Oncology (SIOP) experience. Med Pediatr Oncol. 1999;32(5):317ā€“23.

    PubMedĀ  CASĀ  Google ScholarĀ 

  611. Faria P, Beckwith JB, Mishra K, Zuppan C, Weeks DA, Breslow N, et al. Focal versus diffuse anaplasia in Wilms tumor--new definitions with prognostic significance: a report from the National Wilms Tumor Study Group. Am J Surg Pathol. 1996;20(8):909ā€“20.

    PubMedĀ  CASĀ  Google ScholarĀ 

  612. Vasei M, Moch H, Mousavi A, Kajbafzadeh AM, Sauter G. Immunohistochemical profiling of Wilms tumor: a tissue microarray study. Appl Immunohistochem Mol Morphol. 2008;16(2):128ā€“34.

    PubMedĀ  CASĀ  Google ScholarĀ 

  613. Magro G, Longo FR, Angelico G, Spadola S, Amore FF, Salvatorelli L. Immunohistochemistry as potential diagnostic pitfall in the most common solid tumors of children and adolescents. Acta Histochem. 2015;117(4ā€“5):397ā€“414.

    PubMedĀ  CASĀ  Google ScholarĀ 

  614. Wilms Tumor and Other Childhood Kidney Tumors Treatment (PDQ(R)): Health Professional Version. PDQ Cancer Information Summaries. Bethesda; 2002.

    Google ScholarĀ 

  615. Fuller CE. All things rhabdoid and SMARC: an enigmatic exploration with Dr. Louis P. Dehner. Semin Diagn Pathol. 2016;33(6):427ā€“40.

    PubMedĀ  Google ScholarĀ 

  616. Sebire NJ, Vujanic GM. Paediatric renal tumours: recent developments, new entities and pathological features. Histopathology. 2009;54(5):516ā€“28.

    PubMedĀ  Google ScholarĀ 

  617. Argani P, Perlman EJ, Breslow NE, Browning NG, Green DM, D'Angio GJ, et al. Clear cell sarcoma of the kidney: a review of 351 cases from the National Wilms Tumor Study Group Pathology Center. Am J Surg Pathol. 2000;24(1):4ā€“18.

    PubMedĀ  CASĀ  Google ScholarĀ 

  618. Brownlee NA, Perkins LA, Stewart W, Jackle B, Pettenati MJ, Koty PP, et al. Recurring translocation (10;17) and deletion (14q) in clear cell sarcoma of the kidney. Arch Pathol Lab Med. 2007;131(3):446ā€“51.

    PubMedĀ  Google ScholarĀ 

  619. O'Meara E, Stack D, Lee CH, Garvin AJ, Morris T, Argani P, et al. Characterization of the chromosomal translocation t(10;17)(q22;p13) in clear cell sarcoma of kidney. J Pathol. 2012;227(1):72ā€“80.

    PubMedĀ  CASĀ  Google ScholarĀ 

  620. Furtwaengler R, Reinhard H, Leuschner I, Schenk JP, Goebel U, Claviez A, et al. Mesoblastic nephroma--a report from the Gesellschaft fur Padiatrische Onkologie und Hamatologie (GPOH). Cancer. 2006;106(10):2275ā€“83.

    PubMedĀ  Google ScholarĀ 

  621. Sadler TW. Langmanā€™s medical embryology. 12th ed. Baltimore: Lippincott Williams & Wilkins; 2012.

    Google ScholarĀ 

  622. Reuter V, Al-Ahmadie H, Tickoo S. Urinary bladder, ureter, renal pelvis. In: Mills SE, editor. Histology for pathologists. Philadelphia: LWW; 2012. p. 971ā€“85.

    Google ScholarĀ 

  623. www.meddean.luc.edu/lumen/meded/urology/nlurtdv.htm

  624. Moore KL. The perineum and pelvis. In: Clinically oriented anatomy. 2nd ed. Baltimore: Williams and Wilkin; 1985. p. 298ā€“387.

    Google ScholarĀ 

  625. Tanagho EA. Anatomy of the lower urinary ytacy. In: Walsh PC, et al., editors. Campbellā€™s urology. 5th ed. Phyladelphia: WB Saunders; 1986. p. 46ā€“58.

    Google ScholarĀ 

  626. Bulger RE. The urinary system. In: Weiss L, editor. Cell and tissue biology. 6th ed. Baltimore: Urban and Schwarsenberg; 1988. p. 844ā€“8.

    Google ScholarĀ 

  627. Koss LG. Tumors of the urinary bladder. In: Atlas of tumor pathology, fascicle II. 2nd ed. Washington DC: Armed Forces Institute of pathology; 1975.

    Google ScholarĀ 

  628. Fawcett DW. The urinary system. In: A textbook of histology. 11th ed. Philadelphia: WB Saunders; 1986. p. 787ā€“90.

    Google ScholarĀ 

  629. http://www.cap.org/web/home/protocols-and-guidelines/cancer-reporting-tools/cancer-protocol-templates

  630. Kirkali Z, Chan T, Manoharan M, et al. Bladder cancer: epidemiology, staging, grading and diagnosis. Urology. 2005;66((6) Suppl 1):4ā€“34.

    PubMedĀ  Google ScholarĀ 

  631. Pommer W, Bronder E, Klimpel A, et al. Urothelial cancer at different tumor sites; role of smoking and habitual intake of analgesics and laxatives. Results of the Berlin Urothelial Cancer Study. Nephrol Dial Transplant. 1999;14:2892ā€“7.

    PubMedĀ  CASĀ  Google ScholarĀ 

  632. Zeegers MP, Tan FE, Dorant E, et al. The impact of characteristics of cigarette smoking on urinary tract cancer risk: a meta-analysis of epidemiologic studies. Cancer. 2000;89:630ā€“9.

    PubMedĀ  CASĀ  Google ScholarĀ 

  633. Zeegers MP, Swaen GM, Kant I, et al. Occupational risk factors for male bladder cancer: results from a population based case cohort study in Netherlands. Occup Environ Med. 2001;58:890ā€“6.

    Google ScholarĀ 

  634. Epstein J, Amin MB, Reuter V, et al. 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):1435ā€“48.

    PubMedĀ  CASĀ  Google ScholarĀ 

  635. Humphry PA, Moch H, Cubulla A, et al. The 2016 WHO classification of tumors of the urinary system and male genital organs- part B: prostate and bladder tumors. Eur Urol. 2016;70:106ā€“19.

    Google ScholarĀ 

  636. Amin MB, Smith SC, Reuter VE, et al. Update for the practicing pathologist. The International Consultation on Urologic Disease-European association of urology consultation on bladder cancer. Mod Pathol. 2015;28:612ā€“30.

    PubMedĀ  Google ScholarĀ 

  637. Cheng L, Cheville JC, Neumann RM, et al. Survival of patients with carcinoma in situ of the urinary bladder. Cancer. 1999;85:2469ā€“74.

    PubMedĀ  CASĀ  Google ScholarĀ 

  638. Delbagni G. The management of superficial bladder cancer. Nat Clin Pract Urol. 2007;4:254ā€“60.

    Google ScholarĀ 

  639. Gofrit ON, Pode D, Pizov G, et al. The natural history of bladder carcinoma in situ after initial response to bacillus Calmette-Guerin immunotherapy. Urol Oncol. 2009;27:258ā€“62.

    PubMedĀ  Google ScholarĀ 

  640. Nixon RG, Chang SS, Lafleur BJ, et al. Carcinoma in situ an tumor multifocality predict the risk of practice urethral involvement at radical ecosystems in men with transitional cell carcinoma of the bladder. J Urol. 2002;167(2Pt 1):502ā€“5.

    PubMedĀ  Google ScholarĀ 

  641. Witjes JA. Bladder carcinoma in situ in 2003: state of the art. Eur Urol. 2004;45:142ā€“6.

    PubMedĀ  CASĀ  Google ScholarĀ 

  642. Obermann EC, Junker K, Stoehr R, et al. Frequent genetic alterations in flat urothelial hyperplasia and concomitant papillary bladder cancer as detected by CGH, LOH and FISH analyses. J Pathol. 2003;199:50ā€“7.

    PubMedĀ  CASĀ  Google ScholarĀ 

  643. Van Oers JM, Adam C, Dezinger S, et al. Chromosome 9 deletions are more frequent than FFG3 mutations in flat urothelial hyperplasia of the bladder. Int J Cancer. 2006;119:1212ā€“5.

    PubMedĀ  Google ScholarĀ 

  644. Amin MB, McKenney JK, Paner GP, et al. International Consultation on Urologic Disease-European association of urology consultation on bladder cancer 2012 pathology. Eur Urol. 2013;63:16ā€“35.

    PubMedĀ  Google ScholarĀ 

  645. Hansel DE, Amin MB, Comperat E, et al. A contemporary update on pathology standards for bladder cancer: transurethral resection and radical cystectomy specimens. Eur Urol. 2013;63:321ā€“32.

    PubMedĀ  Google ScholarĀ 

  646. Alsheikh A, Mohamedali Z, Joes E, et al. Compassion of the WHO/ISUP classification and cytokeratin 20 expressions in predicting the behavior of low-grade papillary urothelial tumors. Mod Pathol. 2001;14:267ā€“72.

    PubMedĀ  CASĀ  Google ScholarĀ 

  647. Desai S, Lim SD, Jimenez RE, et al. Relationship of cytokeratin 20 and CD44 protein expression with WHO/ISUP grade in pTa and pT1 papillary urothelial neoplasia. Mod Pathol. 2000;13:135ā€“23.

    Google ScholarĀ 

  648. Fujii Y, Kawakami S, Koga F, et al. Long-term outcome of bladder papillary urothelial neoplasm of low malignant potential. BJU Int. 2003;92:889ā€“62.

    Google ScholarĀ 

  649. Holmang S, Hdelin H, Anderstom C, et al. Recurrence and progression n low grade papillary urothelial tumors. J Urol. 1999;162(3 Pt 1):702ā€“7.

    PubMedĀ  CASĀ  Google ScholarĀ 

  650. Pan CC, Chang YH, Chen KK, et al. Prognostic significances of the 2004 WHO/ISUP classification for prediction of recurrence, progression, and cancer-specific mortality of non-muscle-invasive urothelial tumors of the urinary bladder: a clinicopathologic study of 1, 515 cases. Am J Clin Pathol. 2010;133:788ā€“95.

    PubMedĀ  Google ScholarĀ 

  651. Pich A, Chiusa L, Formicconi A, et al. Biological differences between noninvasive papillary urothelial neoplasms of low malignant potential and low-grade (grade 1) papillary carcinomas of the bladder. Am J Clin Pathol. 2008;25:1528ā€“33.

    Google ScholarĀ 

  652. Kunze E, Schauer A, Schmitt M. Histology and histogenesis of two different types of inverted urothelial papillomas. Cancer. 1983;51:348ā€“58.

    PubMedĀ  CASĀ  Google ScholarĀ 

  653. Picozzi S, Casellato S, Bozzini G, et al. Inverted papilloma of the bladder: a review and an analysis of the recent literature of 365 patients. Urol Oncol. 2013;31:1584ā€“90.

    PubMedĀ  Google ScholarĀ 

  654. Ferlay J, Sorejomataram I, Ervik M, et al. GLOBOCAN 2012; v1.0, Cancer incidence and mortality worldwide: IARC CancerBase No. 11 [internet]. Available from: http://globocan.iarc.fr: International Agency for Research on Cancer. Accessed 19ā€“11ā€“2014.

  655. Tanaka N, Kikuchi E, Kanao K, et al. Metastatic behavior of upper tract urothelial carcinoma after radical nephroureterectomy: association with primary tumor location. Ann Surg Oncol. 2014;21:1038ā€“45.

    PubMedĀ  Google ScholarĀ 

  656. Wallmeroth A, Wagner U, Moch H, et al. Patterns of metastasis in muscle-invasive bladder cancer(pT2ā€“4): An autopsy study on 367 patients. Urol Int. 1999;62:69ā€“75.

    PubMedĀ  CASĀ  Google ScholarĀ 

  657. Denzinger S, Burger M, Fritsche HM, et al. Prognostic value of a histopathological tumor growth pattern at the invasion form of T1G3 urothelial carcinoma of the bladder. Scand J Urol Nephrol. 2009;42:282ā€“7.

    Google ScholarĀ 

  658. Jones EC, Murray SK, Young RH. Cysts and epithelial proliferations of the testicular collecting system (including rete testis). Semin Diagn Pathol. 2000;17:270ā€“93.

    PubMedĀ  CASĀ  Google ScholarĀ 

  659. Amin MB, Trpkov K, Lopez-Beltran A, et al. 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):e20ā€“34.

    PubMedĀ  Google ScholarĀ 

  660. Paner GP, Brown JG, Lapetino S, et al. Diagnostic use of antibody to smoothelin in the recognition of muscularis propria in transurethral resection of urinary bladder tumor (TURBT) specimens. Am J Surg Pathol. 2010;34(6):792ā€“9.

    PubMedĀ  Google ScholarĀ 

  661. Miyamoto H, Sharma RB, Illei PB, et al. Pitfalls in the use of smoothelin to identify muscularis propria invasion by urothelial carcinoma. Am J Surg Pathol. 2010;34(3):418ā€“22.

    PubMedĀ  Google ScholarĀ 

  662. Amin MB, McKenney JK, Paner GP, et al. ICUD-EAU international consultation on bladder cancer 2012: pathology. Eur Urol. 2013;63(1):16ā€“35.

    PubMedĀ  Google ScholarĀ 

  663. Amin M, Tickoo S. Genitourinary. 2nd ed. Salt Lake City: Amirsys; 2016. p. 382ā€“403.

    Google ScholarĀ 

  664. Goebell PJ, Knowles MA. Bladder cancer or bladder cancers? Genetically distinct malignant conditions of the urothelium. Urol on-col. 2010;28:409ā€“28.

    Google ScholarĀ 

  665. Allory Y, Beukers W, Sagrera A, 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;65:360ā€“6.

    PubMedĀ  CASĀ  Google ScholarĀ 

  666. Gunes C, Rudolph KL. The role of telomeres in stem cells and cancer. Cell. 2013;152:390ā€“3.

    PubMedĀ  Google ScholarĀ 

  667. Cancer Genome Atlas Research Network. Comprehensive molecular characterization of urothelial bladder carcinoma. Nature. 2014;507:315ā€“22.

    Google ScholarĀ 

  668. Guo G, Sun X, Chen C, et al. Whole-genome and whole-exome sequencing of bladder cancer identifies frequent alterations in genes involved in sister chromatid cohesion and segregation. Nat Genet. 2013;45:1459ā€“63.

    PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  669. Choi W, Porten S, Kim S, 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):152ā€“65.

    PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  670. Volmar KE, Chan TY, De Marzo AM, et al. Florid von Brunn nests mimicking urothelial carcinoma: a morphologic and immunohistochemical comparison to the nested variant of urothelial carcimona. Am J Surg Pathol. 2003;27:1243ā€“52.

    PubMedĀ  Google ScholarĀ 

  671. Wasco MJ, Daignault S, Bradley D, et al. Nested variant of urothelial carcinoma; a clinicopathologic and immunohistochemical study of 30 pure and mixed cases. Hum Pathol. 2010;41:163ā€“71.

    PubMedĀ  Google ScholarĀ 

  672. Comperat E, Roupret M, Yaxley J, et al. Micropapillary urothelial carcinoma of the urinary bladder: a clinicopathological analysis of 72 cases. Pathology. 2010;42:650ā€“4.

    PubMedĀ  Google ScholarĀ 

  673. Willis DL, Fernandez MI, Dickstein RJ, et al. Clinical outcomes of cT1 micropapillary bladder cancer. J Urol. 2015;193:1129ā€“34.

    PubMedĀ  Google ScholarĀ 

  674. Paner GP, Annaiah C, Gulmann C, et al. Immunohistochemical evaluation of novel and traditional markers associated with urothelial differentiation in a spectrum of variants of urothelial carcinoma of the urinary bladder. Hum Pathol. 2014;45:1473ā€“82.

    PubMedĀ  CASĀ  Google ScholarĀ 

  675. Amin MB, Ro JY, Lee KM, et al. Lymphoepithelioma-like carcinoma of the urinary bladder. Am J Surg Pathol. 1994;18:466ā€“73.

    PubMedĀ  CASĀ  Google ScholarĀ 

  676. Lopez Beltran A, Requena MJ, Montironi R, et al. Plasmacytoid urothelial carcinoma of the bladder. Hum Pathol. 2009;40:1023ā€“8.

    PubMedĀ  Google ScholarĀ 

  677. Mai KT, Park PC, Yazdi HM, et al. Plasmacytoid urothelial carcinoma of the urinary bladder report of severe new cases. Eur Urol. 2006;50:1111ā€“4.

    PubMedĀ  Google ScholarĀ 

  678. Patriarca C, Di Pasquale M, Giunta P, et al. CD-138 positive plasmacytoid urothelial carcinoma of the urinary bladder. Int J Surg Pathol. 2008;16:215ā€“7.

    PubMedĀ  Google ScholarĀ 

  679. Dayyani F, Czemiak BA, Sircar K, et al. Plasmacytoid urothelial carcinoma, a chemosensitive cancer with poor prognosis and peritoneal carcinomatosis. J Urol. 2013;189:1656ā€“61.

    PubMedĀ  Google ScholarĀ 

  680. Keck B, Wash S, Stoehr R, et al. Plasmacytoid variant of bladder cancer defines patients with poor prognosis if treated with cystectomy and adjuvant cisplatin-based chemotherapy. BMC Cancer. 2013;13:71.

    PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  681. Amin MB, Tickoo SK, editors. Urinary bladder in diagnostic pathology: genitourinary. 2nd ed: Amirsys/Elsevier; Salt Lake City, UT; 2016. p. 382ā€“403.

    Google ScholarĀ 

  682. Borhan WN, Cimino-Mathews AM, Montgomery EA, et al. Immunohistochemical differentiation of plasmacytoid urothelial carcinoma from secondary carcinoma involving the bladder. Am J Surg Pathol. 2017;41:1570ā€“5.

    PubMedĀ  Google ScholarĀ 

  683. Lott S, Lopez-Beltran A, Montorini R, et al. Soft tissie tumors of the urinary bladder Part II: malignant neoplasms. Hum Pathol. 2007;38:963ā€“77.

    PubMedĀ  CASĀ  Google ScholarĀ 

  684. Ikegamin H, Iwasaki H, Ohjimi Y, et al. Sarcomatoid carcinoma of the urinary bladder: a clinicopathological and immunohistochemical analysis of 14 patients. Hum Pathol. 2000;31:332ā€“40.

    Google ScholarĀ 

  685. Lopez-Beltram A, Pacelli A, Rothenberg HJ, et al. Carcinosarcoma and and sarcomatoid carcinoma of the bladder: clinicopathologic study of 41 cases. J Urol. 1998;159:1497ā€“503.

    Google ScholarĀ 

  686. Cheng L, Foster SR, McLennan GT, et al. Inflammatory myofibroblastic tumors of the genitourinary tract- single entity or continuum? J Urol. 2008;180:135ā€“40.

    Google ScholarĀ 

  687. Cheng L, Zhang S, Alexander R, et al. Sarcomatoid carcinoma of the urinary bladder: the final common pathway of urothelial carcinoma dedifferentiation. Am J Surg Pathol. 2011;35:e34ā€“6.

    PubMedĀ  Google ScholarĀ 

  688. Samaratunga H, Delahunt B, Egevad L, et al. Pleomorphic giant cell carcinoma of the urinary bladder: an extreme form of tumour de-differentiation. Histopathology. 2016;68:533ā€“40.

    PubMedĀ  Google ScholarĀ 

  689. Samaratunga H, Delahunt B. Recently described and unusual variants of urothelial carcinoma of the urinary bladder. Pathology. 2012;44:407ā€“18.

    PubMedĀ  Google ScholarĀ 

  690. Lopez-Beltran A, Amin MB, Olivera PS, et al. Urothelial carcinoma of the bladder, lipid cell variant: clinicopathologic findings and LOH analysis. Am J Surg Pathol. 2010;34(3):371ā€“6.

    PubMedĀ  Google ScholarĀ 

  691. Kramer MW, Abbas M, Pertschy S, et al. Clear cell variant urothelial carcinoma of the bladder: a case report and review of the literature. Rare Tumors. 2012;4:e48.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  692. Parkin DM. The global burden of urinary bladder cancer. Scand J Urol Neprol Suppl. 2008;218:12ā€“20.

    Google ScholarĀ 

  693. Youssef R, Kapur P, Shariat SF, et al. Prognostic value of apoptotic markers in squamous cell carcinoma of the urinary bladder. BJU Int. 2012;110:961ā€“6.

    PubMedĀ  CASĀ  Google ScholarĀ 

  694. Rambau PF, Chalya PL, Jackson K. Schistosomiasis and urinary bladder cancer in North Western Tanzania: a retrospective review of 185 patients. Inf Agent Cancer. 2013;8:19.

    Google ScholarĀ 

  695. Dematei A, Fernandes R, Soares R, et al. Angiogenesis in Schistosoma haematobium-associated urinary bladder cancer. APMIS. 2017;125:1056ā€“62.

    PubMedĀ  Google ScholarĀ 

  696. Elsobky E, El-Baz M, Gomha M, et al. Prognostic value of angiogenesis in schistosoma-associated squamous cell carcinoma of the urinary bladder. Urology. 2002;60:69ā€“73.

    PubMedĀ  Google ScholarĀ 

  697. Yurdakul T, Avunduk MC, Piskin MM. Pure squamous cell carcinoma after `intravesical BCG treatment. A case report. Urol Int. 2005;74:283ā€“5.

    PubMedĀ  Google ScholarĀ 

  698. Chapman Fredricks JR, Cioffi Lavina M, Accola MA, et al. High risk human papillomavirus DNA detected in primary squamous cell carcinoma of urinary bladder. Arch Pathol Lab Med. 2013;137(8):1088ā€“93.

    PubMedĀ  CASĀ  Google ScholarĀ 

  699. Lagwinski N, Thomas A, Stephenson AJ, et al. Squamous cell carcinoma of the bladder: a clinicopathologic analysis of 45 cases. Am J Surg Pathol. 2007;31:1777ā€“87.

    PubMedĀ  Google ScholarĀ 

  700. Blochin EB, Park KJ, Tickoo SK, et al. Urothelial carcinoma with prominent squamous differentiation in the setting of neurogenic bladder: role of human papillomavirus infection. Mod Pathol. 2012;25:1534ā€“42.

    PubMedĀ  Google ScholarĀ 

  701. Johnson DE, Schoenwald MB, Ayala AG, et al. Squamous cell carcinoma of the bladder. J Urol. 1976;115:542ā€“4.

    PubMedĀ  CASĀ  Google ScholarĀ 

  702. Huang W, Wiiliamson SR, Rao Q, et al. Novel markers of squamous differentiation in the urinary bladder. Hum Pathol. 2013;44(10):1989ā€“97.

    PubMedĀ  CASĀ  Google ScholarĀ 

  703. Scosyrev E, Yao J, Messing E. Urothelial carcinoma versus squamous cell carcinoma of bladder: is survival different with stage adjustment? Urology. 2009;73:822ā€“7.

    PubMedĀ  Google ScholarĀ 

  704. Shokeir AA. Squamous cell carcinoma of the bladder: pathology, diagnosis and treatment. BJU Int. 2004;93:216ā€“20.

    PubMedĀ  CASĀ  Google ScholarĀ 

  705. Muscheck M, Abol-Enein H, Chew K, Moore D 2nd, et al. Comparison of genetic changes in schistosome related transitional and squamous bladder cancers using comparative genomic hybridization. Carcinogensis. 2000;21:1721ā€“6.

    CASĀ  Google ScholarĀ 

  706. Gonzalez-Zulueta M, Shibata A, Ohneseit PF, et al. High frequency of chromosome 9p allelic loss and CDKN2 tumor suppressor gene alterations in squamous cell carcinoma of the bladder. J Natl Cancer Inst. 1995;87:1383ā€“93.

    PubMedĀ  CASĀ  Google ScholarĀ 

  707. Aly MS, Khaled HM. Chromosomal aberrations in early-stage bilharzia! bladder cancer. Cancer Genet Cytogenet. 2002;132:41ā€“5.

    PubMedĀ  CASĀ  Google ScholarĀ 

  708. El-Rifai W, Kamel D, Larramendy ML, et al. DNA copy number changes in Schistoso ma-associated and non-Schistosoma-associated bladder cancer. Am J Pathol. 2000;156:871ā€“8.

    PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  709. El-Sebai I, Sherif M, El Bolkainy MN, et al. Verrucose squamous carcinoma of bladder. Urology. 1974;4:407ā€“10.

    PubMedĀ  CASĀ  Google ScholarĀ 

  710. Batta AG, Engen DE, Reiman HM, et al. Intravesical condyloma acuminalum with progression to verrucous carcinoma. Urology. 1990;36:457ā€“64.

    PubMedĀ  CASĀ  Google ScholarĀ 

  711. Mahran MR, El-Baz M. Verrocous carcinoma of the bilharzial bladder. Impact of invasiveness on survival. Scand J Urol Nephrol. 1993;27:189ā€“1992.

    PubMedĀ  CASĀ  Google ScholarĀ 

  712. Grignon D. Tumors of the urinary bladder. In: Amin M, Grignon D, Srigley J, Eble J, editors. Urological pathology. 1st ed. Philadelphia: Lippincott, Williams and Wilkins; 2015. p. 345-B.

    Google ScholarĀ 

  713. Guo CC, Fine SW, Epstein JI. Noninvasive squamous lesions in the urinary bladder: a clinicopathologic analysis of 29 cases. Am J Surg Pathol. 2016;30:883ā€“91.

    Google ScholarĀ 

  714. Ploeg M, Aben KK, Hulsbergen-van deKaa CA, et al. Clinical epidemiology of nonurothelial bladder cancer: analysis of the Netherlands Cancer Registry. J Urol. 2010;183:915ā€“20.

    PubMedĀ  Google ScholarĀ 

  715. Gordon NS, Sinclair RA, Snow RM. Pelvic lipomatosis with cystitis cyslica, cystitis glandularis and adenocarcinoma of the bladder: first reported case. Aust N Z J Surg. 1990;60:229ā€“32.

    PubMedĀ  CASĀ  Google ScholarĀ 

  716. Engel RM, Wilkinson HA. Bladder exstrophy. J Urol. 1970;104:699ā€“704.

    PubMedĀ  CASĀ  Google ScholarĀ 

  717. Grignon DJ, Ro JY, Ayala AG, Johnson DE, et al. Primary adenocarcinoma of the urinary bladder. A clinicopathologic analysis of 72 cases. Cancer. 1991;67:2165ā€“72.

    PubMedĀ  CASĀ  Google ScholarĀ 

  718. Wang HL, Lu OW, Yerian LM, et al. Immunohistochemical distinction between primary adenocarcinoma of the bladder and secondary colorectal adenocarcinoma. Am J Surg Pathol. 2001;25:1360ā€“7.

    Google ScholarĀ 

  719. Amin MB, Edge S, Greene F, et al. AJCC Cancer staging manual. 8th ed. New York: Springer-Verlag; 2017.

    Google ScholarĀ 

  720. Cheng L, et al. Villous adenoma of the urinary tract: a report of 23 cases, including 8 with coexistent adenocarcinoma. Am J Surg Pathol. 1999;23:764ā€“71.

    PubMedĀ  CASĀ  Google ScholarĀ 

  721. Lane Z, Hansel DE, Epstein JI. Immunohistochemical expression of prostatic antigens in adenocarcinoma and villous adenoma of the urinary bladder. Am J Surg Pathol. 2008;32:1322ā€“6.

    PubMedĀ  Google ScholarĀ 

  722. Begg R. The colloid adenocarcinoma of the bladder vault arising from the epithelium of the urachal canal: with a critical survey of the tumors of the urachus. Br J Surg. 1931;18:422ā€“64.

    Google ScholarĀ 

  723. Amin MB, Smith SC, Eble JN, Rao P, et al. Glandular neoplasms of the urachus: a report of 55 cases emphasizing mucinous cystic tumors with proposed classification. Am J Surg Pathol. 2014;38:1033ā€“45.

    PubMedĀ  Google ScholarĀ 

  724. Gopalan A, Sharp OS, Fine SW, et al. UraĀ·chal carcinoma: a clinicopathologic analysis of 24 cases with outcome correlation. Am J Surg Pathol. 2009;3:659ā€“68.

    Google ScholarĀ 

  725. Paner GP, McKenney JK, Barkan GA, et al. Immunohistochemical analysis in a morphologic spectrum of urachal epithelial neoplasms: diagnostic implications and pitfalls. Am J Surg Pathol. 2011;35:787ā€“98.

    PubMedĀ  Google ScholarĀ 

  726. Wright JL, Porter MP, Li CJ, et al. Differences in survival among patients with urachal and nonurachal adenocarcinomas of the bladder. Cancer. 2006;107:721ā€“8.

    PubMedĀ  Google ScholarĀ 

  727. Sheldon CA, Clayman RV, Gonzalez R, et al. Malignant urachal lesions. J Urol. 1984;131:1ā€“8.

    PubMedĀ  CASĀ  Google ScholarĀ 

  728. Sirintrapun SJ, Ward M, Woo J, Cimic A. High-stage urachal adenocarcinoma can be associated with microsatellite instability and KRAS mutations. Hum Pathol. 2014;45:327ā€“30.

    PubMedĀ  CASĀ  Google ScholarĀ 

  729. Young RH, Clement PB. Mulle rianosis of the urinary bladder. Mod Pathol. 1996;9:731ā€“7.

    PubMedĀ  CASĀ  Google ScholarĀ 

  730. Oliva E, Amin MB, Jimenez R, et al. Clear cell carcinoma of the urinary bladder: a report and comparison of four tumors of mullerian origin and nine of probable urothelial origin with discussion of histogenesis and diagnostic problems. Am J Surg Pathol. 2002;6:190ā€“7.

    Google ScholarĀ 

  731. Al-Izzi MS, Horton LW, Kelleher J, et al. Malignant transformation in endometriosis of the urinary bladder. Histopathology. 1989;14:191ā€“8.

    PubMedĀ  CASĀ  Google ScholarĀ 

  732. Allen D, Oā€™Brien T, Pingle P, Chandra A. Endometrioid adenocarcinoma of the bladder. Histopathology. 2005;46:232ā€“3.

    PubMedĀ  CASĀ  Google ScholarĀ 

  733. Young RH, Clement PB. Mullerianosis of the urinary bladder. Mod Pathol. 1996;9:731ā€“7.

    PubMedĀ  CASĀ  Google ScholarĀ 

  734. Jones TD, Kemek KM, Yang XJ, et al. Thyroid transcription factor 1expression in small cell carcinoma of the urinary bladder: an immunohistochemical profile of 44 cases. Hum Pathol. 2005;36:718ā€“23.

    PubMedĀ  CASĀ  Google ScholarĀ 

  735. Thompson S, Cioffi-Lavina M, Chapman-Fredricks J, et al. Differential diagnosis: poorly differentiated urothelial carcinoma (lacks neuroendocrine markers and pankeratin expression is non-dot like fashion), Distinction of high-grade neuroendocrine carcinoma/small cell carcinoma from conventional urothelial carcinoma of urinary bladder: an immunohistochemical approach. Appl Immunohistochem Mol Morphol. 2011;19(5):395ā€“9.

    PubMedĀ  CASĀ  Google ScholarĀ 

  736. Lyer G, Al-Ahmadie H, Schultz N, et al. Prevalence and co-occurrence of actionable genomic alterations in high-grade bladder cancer. J Clin Oneal. 2013;31:3133ā€“40.

    Google ScholarĀ 

  737. Chen YB, Epstein JI. Primary carcinoid of the urinary bladder and prostatic urethra: a clinicopathologic study of 6 cases. Am J Surg Pathol. 2011;35:442.

    PubMedĀ  CASĀ  Google ScholarĀ 

  738. Martucci VL, et al. Association of urinary bladder paraganglioma with germ-line mutations in the SDHB and VHL genes. Urol Oncol. 2015;33(4):167 e13ā€“20.

    Google ScholarĀ 

  739. So JS. Epstein JI GATA3 expression in paragangliomas;a pitfall potentially leading to misdiagnosis of urothelial carcinoma. Mod Pathol. 2013;26:1365ā€“70.

    PubMedĀ  CASĀ  Google ScholarĀ 

  740. Beilan JA, Lawton A, Hajdenberg J, Rosser CJ. Pheochromocytoma of the urinary bladder; a systematic review of the contemporary literature. BMC Urol. 2013;13:22.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  741. Mason EF, Sadow PM, Wagner AJ, et al. Identification of succinate dehydrogenase deficient bladder paragangliomas. Am J Surg Pathol. 2013;37:1612ā€“8.

    PubMedĀ  Google ScholarĀ 

  742. Venyo AK. Melanoma of the urinary bladder: a review of the literature. Surg Res Pract. 2014;2014:605802.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  743. Ainwaorth AM, Clark WH, Mastrangelo M, et al. Primary malignant melanoma of the urinary bladder. Cancer. 1976;37:1828ā€“36.

    Google ScholarĀ 

  744. Kim J, McCarthy SW, Thompson JF, Pupo GM, Vonthethoff L, Nash P, et al. Cellular blue naevus involving the urinary bladder. Pathology. 2012;44:664ā€“8.

    PubMedĀ  Google ScholarĀ 

  745. Patel P, Gotto G, Kavanagh A, et al. Urinary bladder melanosis associated with urothelial dysplasia and invasive urothelial carcinoma: a report of two cases. Anal Quant Cytopathol Histpathol. 2013;35:294ā€“300.

    PubMedĀ  Google ScholarĀ 

  746. Paner GP, McKenny JK, Barkan GA, et al. Rhabdomyosarcoma of the urinary bladder in adults: predilection for alveolar morphology with anaplasia and significant morphologic overlap with small cell carcinoma. Am J Surg Pathol. 2008;32:1022ā€“08.

    PubMedĀ  Google ScholarĀ 

  747. Mills SE, Bova GS, Wick MR, et al. Leiomyosarcoma of the urinary bladder. A clinicopathologic and immunohistochemical study of 15 cases. Am J Surg Pathol. 1989;113:480ā€“9.

    Google ScholarĀ 

  748. Williams S, Romaguera R, Kava B. Angiosarcoma of the bladder. Case report and review of the literature. Scientific World J. 2008;8:508ā€“11.

    Google ScholarĀ 

  749. Coffin CM, Watterson J, Priest JR, et al. Extrapulmonary inflammatory myofibroblastic tumor (inflammatory pseudotumor). A clinicopathological and immunohistochemical stydy of 84 cases. Am J Surg Pathol. 1995;19:859ā€“72.

    PubMedĀ  CASĀ  Google ScholarĀ 

  750. Coffin CM, Hornick JL, Fletcher CD. Inflammatory myofibroblastic tumor: comparison of clinicopathologic, histologic, and immunohistochemical features including ALK expression in atypical and aggressive cases. Am J Surg Pathol. 2007;31:509ā€“20.

    PubMedĀ  Google ScholarĀ 

  751. Sukov WR, Cheville JC, Amin MB, et al. Perivascular epithelioid cell tumor (PEComa) of the urinary bladder: report of 3 cases and review of the literature. Am J Surg Pathol. 2009;33:304ā€“8.

    PubMedĀ  Google ScholarĀ 

  752. Amin M, Tickoo S. Genitourinary. 2nd ed. Salt Lake City: Amirsys; 2016. p. 470ā€“7.

    Google ScholarĀ 

  753. Amin M, Tickoo S. Genitourinary. 2nd ed. Salt Lake City: Amirsys; 2016. p. 482ā€“5.

    Google ScholarĀ 

  754. Sharma KV, Venkatesan AM, Swerdlow D, DaSilva D, Beck A, Jain N, Wood BJ. Image-guided adrenal and renal biopsy. Tech Vasc Interv Radiol. 2010;13(2):100ā€“9.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  755. Villelli NW, Jayanti MK, Zynger DL. Use and usefulness of adrenal core biopsies without FNA or on-site evaluation of adequacy: a study of 204 cases for a 12-year period. Am J Clin Pathol. 2012;137(1):124ā€“31.

    PubMedĀ  Google ScholarĀ 

  756. Welch TJ, Sheedy PF 2nd, Stephens DH, et al. Percutaneous adrenal biopsy: review of a 10-year experience. Radiology. 1994;193:341ā€“4.

    PubMedĀ  CASĀ  Google ScholarĀ 

  757. Lumachi F, Borsato S, Tregnaghi A, et al. CT-scan, MRI and image-guided FNA cytology of incidental adrenal masses. Eur J Surg Oncol. 2003;29:689ā€“92.

    PubMedĀ  CASĀ  Google ScholarĀ 

  758. Quayle FJ, Spitler JA, Pierce RA, Lairmore TC, Moley JF, Brunt LM. Needle biopsy of incidentally discovered adrenal masses is rarely informative and potentially hazardous. Surgery. 2007;142(4):497ā€“502.

    PubMedĀ  Google ScholarĀ 

  759. DeWitt J, Alsatie M, LeBlanc J, McHenry L, Sherman S. Endoscopic ultrasound-guided fine-needle aspiration of left adrenal gland masses. Endoscopy. 2007;39(1):65ā€“71.

    PubMedĀ  CASĀ  Google ScholarĀ 

  760. Ball MW, Hemal AK, Allaf ME. International Consultation on Urological Diseases and European Association of Urology international consultation on minimally invasive surgery in urology: laparoscopic and robotic adrenalectomy. BJU Int. 2017;119(1):13ā€“21.

    PubMedĀ  Google ScholarĀ 

  761. Carr AA, Wang TS. Minimally invasive adrenalectomy. Surg Oncol Clin N Am. 2016;25(1):139ā€“52.

    PubMedĀ  Google ScholarĀ 

  762. Protocol for the examination of specimens from patients with carcinoma of the adrenal gland. College of American Pathologists. http://www.cap.org/ShowProperty?nodePath=/UCMCon/ContributionFolders/WebContent/pdf/cp-adrenal-17protocol-4000.pdf. Accessed 17 July 2017.

  763. Protocol for the examination of specimens from patients with neuroblastoma. http://www.cap.org/ShowProperty?nodePath=/UCMCon/ContributionFolders/WebContent/pdf/neuroblastoma-16.pdf. Accessed 17 July 2017.

  764. Barzon L, Sonino N, Fallo F, Palu G, Boscaro M. Prevalence and natural history of adrenal incidentalomas. Eur J Endocrinol. 2003;149(4):273ā€“85.

    PubMedĀ  CASĀ  Google ScholarĀ 

  765. Lerario AM, Moraitis A, Hammer GD. Genetics and epigenetics of adrenocortical tumors. Mol Cell Endocrinol. 2014;386(1ā€“2):67ā€“84.

    PubMedĀ  CASĀ  Google ScholarĀ 

  766. Shastri C, Rana C, Kumari N, Agarwal G, Krishnani N. Bilateral adrenocortical oncocytoma with bilateral myelolipomatous metaplasia. Endocr Pathol. 2012;23(2):112ā€“4.

    PubMedĀ  Google ScholarĀ 

  767. Brown FM, Gaffey TA, Wold LE, Lloyd RV. Myxoid neoplasms of the adrenal cortex: a rare histologic variant. Am J Surg Pathol. 2000;24(3):396ā€“401.

    PubMedĀ  CASĀ  Google ScholarĀ 

  768. Raparia K, Ayala AG, Sienko A, Zhai QJ, Ro JY. Myxoid adrenal cortical neoplasms. Ann Diagn Pathol. 2008;12(5):344ā€“8.

    PubMedĀ  Google ScholarĀ 

  769. Macadam RF. Black adenoma of the human adrenal cortex. Cancer. 1971;27(1):116ā€“9.

    PubMedĀ  CASĀ  Google ScholarĀ 

  770. Kameyama K, Takami H. Pigmented granules in functional black adenoma of the adrenal gland: a histochemical and ultrastructural study. Endocr Pathol. 1999;10(4):353ā€“7.

    PubMedĀ  Google ScholarĀ 

  771. Weiss LM. Comparative histologic study of 43 metastasizing and nonmetastasizing adrenocortical tumors. Am J Surg Pathol. 1984;8(3):163ā€“9.

    PubMedĀ  CASĀ  Google ScholarĀ 

  772. Aubert S, Wacrenier A, Leroy X, Devos P, Carnaille B, Proye C, Wemeau JL, Lecomte-Houcke M, Leteurtre E. Weiss system revisited: a clinicopathologic and immunohistochemical study of 49 adrenocortical tumors. Am J Surg Pathol. 2002;26(12):1612ā€“9.

    PubMedĀ  Google ScholarĀ 

  773. Lloyd RV. Adrenal cortical tumors, pheochromocytomas and paragangliomas. Mod Pathol. 2011;24(Suppl 2):S58ā€“65.

    PubMedĀ  CASĀ  Google ScholarĀ 

  774. Busam KJ, Iversen K, Coplan KA, Old LJ, Stockert E, Chen YT, McGregor D, Jungbluth A. Immunoreactivity for A103, an antibody to melan-A (Mart-1), in adrenocortical and other steroid tumors. Am J Surg Pathol. 1998;22(1):57ā€“63.

    PubMedĀ  CASĀ  Google ScholarĀ 

  775. Jorda M, De MB, Nadji M. Calretinin and inhibin are useful in separating adrenocortical neoplasms from pheochromocytomas. Appl Immunohistochem Mol Morphol. 2002;10(1):67ā€“70.

    PubMedĀ  Google ScholarĀ 

  776. Loy TS, Phillips RW, Linder CL. A103 immunostaining in the diagnosis of adrenal cortical tumors: an immunohistochemical study of 316 cases. Arch Pathol Lab Med. 2002;126(2):170ā€“2.

    PubMedĀ  Google ScholarĀ 

  777. McCluggage WG, Burton J, Maxwell P, Sloan JM. Immunohistochemical staining of normal, hyperplastic, and neoplastic adrenal cortex with a monoclonal antibody against alpha inhibin. J Clin Pathol. 1998;51(2):114ā€“6.

    PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  778. Pelkey TJ, Frierson HF Jr, Mills SE, Stoler MH. The alpha subunit of inhibin in adrenal cortical neoplasia. Mod Pathol. 1998;11(6):516ā€“24.

    PubMedĀ  CASĀ  Google ScholarĀ 

  779. Zhang PJ, Genega EM, Tomaszewski JE, Pasha TL, LiVolsi VA. The role of calretinin, inhibin, melan-A, BCL-2, and C-kit in differentiating adrenal cortical and medullary tumors: an immunohistochemical study. Mod Pathol. 2003;16(6):591ā€“7.

    PubMedĀ  Google ScholarĀ 

  780. Zhang H, Bu H, Chen H, Wei B, Liu W, Guo J, Li F, Liao D, Tang Y, Zhang Z. Comparison of immunohistochemical markers in the differential diagnosis of adrenocortical tumors: immunohistochemical analysis of adrenocortical tumors. Appl Immunohistochem Mol Morphol. 2008;16(1):32ā€“9.

    PubMedĀ  CASĀ  Google ScholarĀ 

  781. Sbiera S, Schmull S, Assie G, Voelker HU, Kraus L, Beyer M, Ragazzon B, Beuschlein F, Willenberg HS, Hahner S, Saeger W, Bertherat J, Allolio B, Fassnacht M. High diagnostic and prognostic value of steroidogenic factor-1 expression in adrenal tumors. J Clin Endocrinol Metab. 2010;95(10):E161ā€“71.

    PubMedĀ  CASĀ  Google ScholarĀ 

  782. Schrƶder S, Padberg BC, Achilles E, Holl K, Dralle H, Klƶppel G. Immunocytochemistry in adrenocortical tumours: a clinicomorphological study of 72 neoplasms. Virchows Arch A Pathol Anat Histopathol. 1992;420(1):65ā€“70.

    PubMedĀ  Google ScholarĀ 

  783. Donato DP, Johnson MT, Yang XJ, Zynger DL. Expression of carbonic anhydrase IX in genitourinary and adrenal tumours. Histopathology. 2011;59(6):1229ā€“39.

    PubMedĀ  Google ScholarĀ 

  784. Papotti M, Duregon E, Volante M, McNicol AM. Pathology of the adrenal cortex: a reappraisal of the past 25 years focusing on adrenal cortical tumors. Endocr Pathol. 2014;25(1):35ā€“48.

    PubMedĀ  CASĀ  Google ScholarĀ 

  785. Zeiger MA, Thompson GB, Duh QY, Hamrahian AH, Angelos P, Elaraj D, Fishman E, Kharlip J, American Association of Clinical Endocrinologists; American Association of Endocrine Surgeons. The American Association of Clinical Endocrinologists and American Association of Endocrine Surgeons medical guidelines for the management of adrenal incidentalomas. Endocr Pract. 2009;15(Suppl 1):1ā€“20.

    PubMedĀ  Google ScholarĀ 

  786. Zeiger MA, Thompson GB, Duh QY, Hamrahian AH, Angelos P, Elaraj D, Fishman E, Kharlip J, American Association of Clinical Endocrinologists; American Association of Endocrine Surgeons. American Association of Clinical Endocrinologists and American Association of Endocrine Surgeons Medical Guidelines for the Management of Adrenal Incidentalomas: executive summary of recommendations. Endocr Pract. 2009;15(5):450ā€“3.

    PubMedĀ  Google ScholarĀ 

  787. Kim Y, Margonis GA, Prescott JD, Tran TB, Postlewait LM, Maithel SK, Wang TS, Glenn JA, Hatzaras I, Shenoy R, Phay JE, Keplinger K, Fields RC, Jin LX, Weber SM, Salem A, Sicklick JK, Gad S, Yopp AC, Mansour JC, Duh QY, Seiser N, Solorzano CC, Kiernan CM, Votanopoulos KI, Levine EA, Poultsides GA, Pawlik TM. Curative surgical resection of adrenocortical carcinoma: determining long-term outcome based on conditional disease-free probability. Ann Surg. 2017;265(1):197ā€“204.

    PubMedĀ  Google ScholarĀ 

  788. Choi YM, Kwon H, Jeon MJ, Sung TY, Hong SJ, Kim TY, Kim WB, Shong YK, Lee JL, Song DE, Kim WG. Clinicopathological features associated with the prognosis of Pat812, ients with adrenal cortical carcinoma: usefulness of the Ki-67 index. Medicine (Baltimore). 2016;95(21):e3736.

    Google ScholarĀ 

  789. Margonis GA, Amini N, Kim Y, Tran TB, Postlewait LM, Maithel SK, Wang TS, Evans DB, Hatzaras I, Shenoy R, Phay JE, Keplinger K, Fields RC, Moses LE, Weber SM, Salem A, Sicklick JK, Gad S, Yopp AC, Mansour JC, Duh QY, Seiser N, Solorzano CC, Kiernan CM, Votanopoulos KI, Levine EA, Poultsides GA, Pawlik TM. Incidence of perioperative complications following resection of adrenocortical carcinoma and its association with long-term survival. World J Surg. 2016;40(3):706ā€“14.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  790. Loncar Z, Djukic V, Zivaljevic V, Pekmezovic T, Diklic A, Tatic S, Dundjerovic D, Olujic B, Slijepcevic N, Paunovic I. Survival and prognostic factors for adrenocortical carcinoma: a single institution experience. BMC Urol. 2015;15:43.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  791. Calissendorff J, Calissendorff F, Falhammar H. Adrenocortical cancer: mortality, hormone secretion, proliferation and urine steroids - experience from a single Centre spanning three decades. BMC Endocr Disord. 2016;16:15.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  792. Sabaretnam M, Mishra A, Agarwal G, Agarwal A, Verma AK, Mishra SK. Adrenocortical carcinoma in children and adults: two decades experience in a single institution. Indian J Cancer. 2016;53(2):317ā€“21.

    PubMedĀ  CASĀ  Google ScholarĀ 

  793. Wang C, Sun Y, Wu H, Zhao D, Chen J. Distinguishing adrenal cortical carcinomas and adenomas: a study of clinicopathological features and biomarkers. Histopathology. 2014;64(4):567ā€“76.

    PubMedĀ  Google ScholarĀ 

  794. Duregon E, Cappellesso R, Maffeis V, Zaggia B, Ventura L, Berruti A, Terzolo M, Fassina A, Volante M, Papotti M. Validation of the prognostic role of the ā€œHelsinki scoreā€ in 225 cases of adrenocortical carcinoma. Hum Pathol. 2017;62:1ā€“7.

    PubMedĀ  Google ScholarĀ 

  795. de Krijger RR, Papathomas TG. Adrenocortical neoplasia: evolving concepts in tumorigenesis with an emphasis on adrenal cortical carcinoma variants. Virchows Arch. 2012;460(1):9ā€“18.

    PubMedĀ  Google ScholarĀ 

  796. Sangoi AR, Fujiwara M, West RB, Montgomery KD, Bonventre JV, Higgins JP, Rouse RV, Gokden N, McKenney JK. Immunohistochemical distinction of primary adrenal cortical lesions from metastatic clear cell renal cell carcinoma: a study of 248 cases. Am J Surg Pathol. 2011;35(5):678ā€“86.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  797. Renshaw AA, Granter SR. A comparison of A103 and inhibin reactivity in adrenal cortical tumors: distinction from hepatocellular carcinoma and renal tumors. Mod Pathol. 1998;11(12):1160ā€“4.

    PubMedĀ  CASĀ  Google ScholarĀ 

  798. Cho EY, Ahn GH. Immunoexpression of inhibin alpha-subunit in adrenal neoplasms. Appl Immunohistochem Mol Morphol. 2001;9(3):222ā€“8.

    PubMedĀ  CASĀ  Google ScholarĀ 

  799. Elmoneim HM, Samaka RM, Ali H. Diagnostic role of inhibin Ī±-subunit and inhibin/activin Ī²-subunit in adrenal cortical and medullary tumors in Egyptian patients. Appl Immunohistochem Mol Morphol. 2012;20(5):462ā€“9.

    PubMedĀ  Google ScholarĀ 

  800. Sangoi AR, McKenney JK. A tissue microarray-based comparative analysis of novel and traditional immunohistochemical markers in the distinction between adrenal cortical lesions and pheochromocytoma. Am J Surg Pathol. 2010;34(3):423ā€“32.

    PubMedĀ  Google ScholarĀ 

  801. Weissferdt A, Phan A, Suster S, Moran CA. Adrenocortical carcinoma: a comprehensive immunohistochemical study of 40 cases. Appl Immunohistochem Mol Morphol. 2014;22(1):24ā€“30.

    PubMedĀ  CASĀ  Google ScholarĀ 

  802. Perrino CM, Ho A, Dall CP, Zynger DL. Utility of GATA3 in the differential diagnosis of pheochromocytoma. Histopathology. 2017;71(3):475ā€“9. https://doi.org/10.1111/his.13229. Epub 2017 Jun 5.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  803. Duregon E, Volante M, Bollito E, Goia M, Buttigliero C, Zaggia B, Berruti A, Scagliotti GV, Papotti M. Pitfalls in the diagnosis of adrenocortical tumors: a lesson from 300 consultation cases. Hum Pathol. 2015;46(12):1799ā€“807.

    PubMedĀ  Google ScholarĀ 

  804. Margonis GA, Kim Y, Prescott JD, Tran TB, Postlewait LM, Maithel SK, Wang TS, Evans DB, Hatzaras I, Shenoy R, Phay JE, Keplinger K, Fields RC, Jin LX, Weber SM, Salem A, Sicklick JK, Gad S, Yopp AC, Mansour JC, Duh QY, Seiser N, Solorzano CC, Kiernan CM, Votanopoulos KI, Levine EA, Poultsides GA, Pawlik TM. Adrenocortical carcinoma: impact of surgical margin status on long-term outcomes. Ann Surg Oncol. 2016;23(1):134ā€“41.

    PubMedĀ  Google ScholarĀ 

  805. Simon G, Pattou F, MiralliĆ© E, Lifante JC, NominĆ© C, Arnault V, de Calan L, Caillard C, Carnaille B, Brunaud L, Laplace N, Caiazzo R, Blanchard C. Surgery for recurrent adrenocortical carcinoma: a multicenter retrospective study. Surgery. 2017;161(1):249ā€“56.

    PubMedĀ  Google ScholarĀ 

  806. Berruti A, Grisanti S, Pulzer A, Claps M, Daffara F, Loli P, Mannelli M, Boscaro M, Arvat E, Tiberio G, Hahner S, Zaggia B, Porpiglia F, Volante M, Fassnacht M, Terzolo M. Long-term outcomes of adjuvant mitotane therapy in patients with radically resected adrenocortical carcinoma. J Clin Endocrinol Metab. 2017;102(4):1358ā€“65.

    PubMedĀ  Google ScholarĀ 

  807. PatĆ³cs A, KarĆ”di E, TĆ³th M, Varga I, SzĆ¼cs N, Balogh K, Majnik J, GlĆ”z E, RĆ”cz K. Clinical and biochemical features of sporadic and hereditary phaeochromocytomas: an analysis of 41 cases investigated in a single endocrine centre. Eur J Cancer Prev. 2004;13(5):403ā€“9.

    PubMedĀ  Google ScholarĀ 

  808. Kulis T, Knezevic N, Pekez M, Kastelan D, Grkovic M, Kastelan Z. Laparoscopic adrenalectomy: lessons learned from 306 cases. J Laparoendosc Adv Surg Tech A. 2012;22(1):22ā€“6.

    PubMedĀ  Google ScholarĀ 

  809. Amar L, Bertherat J, Baudin E, Ajzenberg C, Bressac-de Paillerets B, Chabre O, Chamontin B, Delemer B, Giraud S, Murat A, Niccoli-Sire P, Richard S, Rohmer V, Sadoul JL, Strompf L, Schlumberger M, Bertagna X, Plouin PF, Jeunemaitre X, Gimenez-Roqueplo AP. Genetic testing in pheochromocytoma or functional paraganglioma. J Clin Oncol. 2005;23(34):8812ā€“8.

    PubMedĀ  CASĀ  Google ScholarĀ 

  810. Jiang S, Dahia PL. Minireview: the busy road to pheochromocytomas and paragangliomas has a new member, TMEM127. Endocrinology. 2011;152(6):2133ā€“40.

    PubMedĀ  CASĀ  Google ScholarĀ 

  811. Guerrero MA, Schreinemakers JM, Vriens MR, et al. Clinical spectrum of pheochromocytoma. J Am Coll Surg. 2009;209:727.

    PubMedĀ  Google ScholarĀ 

  812. Young WF Jr, Maddox DE. Spells: in search of a cause. Mayo Clin Proc. 1995;70:757.

    PubMedĀ  Google ScholarĀ 

  813. Manger WM, Gifford RW. Pheochromocytoma. J Clin Hypertens (Greenwich). 2002;4:62.

    Google ScholarĀ 

  814. Baguet JP, Hammer L, Mazzuco TL, et al. Circumstances of discovery of phaeochromocytoma: a retrospective study of 41 consecutive patients. Eur J Endocrinol. 2004;150:681.

    PubMedĀ  CASĀ  Google ScholarĀ 

  815. Lu Y, Li P, Gan W, Zhao X, Shen S, Feng W, Xu Q, Bi Y, Guo H, Zhu D. Clinical and pathological characteristics of hypertensive and normotensive adrenal pheochromocytomas. Exp Clin Endocrinol Diabetes. 2016;124(6):372ā€“9.

    PubMedĀ  CASĀ  Google ScholarĀ 

  816. Alsabeh R, Mazoujian G, Goates J, Mederios LJ, Weiss LM. Adrenal cortical tumors clinically mimicking pheochromocytoma. Am J Clin Pathol. 1995;104(4):382ā€“90.

    PubMedĀ  CASĀ  Google ScholarĀ 

  817. Ivsic T, Komorowski RA, Sudakoff GS, Wilson SD, Datta MW. Adrenal cortical adenoma with adrenalin-type neurosecretory granules clinically mimicking a pheochromocytoma. Arch Pathol Lab Med. 2002;126(12):1530ā€“3.

    PubMedĀ  Google ScholarĀ 

  818. Simon S, Tƶtsch M, Schmidlin F, Iselin CE, Meier CA. Adrenal cortical phaeochromocytoma: a case report of a rare entity. Exp Clin Endocrinol Diabetes. 2003;111(2):111ā€“4.

    PubMedĀ  CASĀ  Google ScholarĀ 

  819. Horchani A, Nouira Y, Nouira K, et al. Hydatid cyst of the adrenal gland: a clinical study of six cases. Sci World J. 2006;6:2420ā€“5.

    Google ScholarĀ 

  820. Lee JA, Zarnegar R, Shen WT, Kebebew E, Clark OH, Duh QY. Adrenal incidentaloma, borderline elevations of urine or plasma metanephrine levels, and the ā€œsubclinicalā€ pheochromocytoma. Arch Surg. 2007;142(9):870ā€“3.

    PubMedĀ  CASĀ  Google ScholarĀ 

  821. Kiriakopoulos A, Papaioannou D, Linos D. Adrenal cortical oncocytoma mimicking pheochromocytoma. Hormones (Athens). 2011;10(1):76ā€“9.

    Google ScholarĀ 

  822. Perrino CM, Prall DN, Calomeni EP, Nadasdy T, Zynger DL. Ultrastructural findings in adrenal cortical adenomas clinically mimicking pheochromocytoma: a comparison with other adrenal tumors and tissue preparation techniques. Ultrastruct Pathol. 2012;36(5):287ā€“93.

    PubMedĀ  Google ScholarĀ 

  823. Morse MO, Schwartz FL, Zynger DL. First report of adrenal cortical endothelial (vascular) cyst mimicking phaeochromocytoma. Pathology. 2014;46(4):364ā€“5.

    PubMedĀ  Google ScholarĀ 

  824. Barmpari ME, Savvidis C, Dede AD, Markogiannakis H, Dikoglou C, Xekouki P, Stratakis CA, Manouras A, Malaktari-Skarantavou S. Adrenal malignant melanoma masquerading as a pheochromocytoma in a patient with a history of a multifocal papillary and medullary thyroid carcinoma. Hormones (Athens). 2016;15(2):283ā€“90.

    Google ScholarĀ 

  825. Heimburger C, Averous G, Charlin E, Lang H, Kurtz JE, Imperiale A. Adrenal metastasis of a poorly differentiated adenocarcinoma mimicking a pheochromocytoma on 18F-FDOPA PET/CT. Clin Nucl Med. 2016;41(9):691ā€“2.

    PubMedĀ  Google ScholarĀ 

  826. Humphrey R, Gray D, Pautler S, Davies W. Laparoscopic compared with open adrenalectomy for resection of pheochromocytoma: a review of 47 cases. Can J Surg. 2008;51(4):276ā€“80.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  827. Kasahara T, Nishiyama T, Takahashi K. Laparoscopic adrenalectomy for pheochromocytoma: evaluation of experience and strategy at a single institute. BJU Int. 2009;103(2):218ā€“22.

    PubMedĀ  Google ScholarĀ 

  828. Feng F, Zhu Y, Wang X, Wu Y, Zhou W, Jin X, Zhang R, Sun F, Kasoma Z, Shen Z. Predictive factors for malignant pheochromocytoma: analysis of 136 patients. J Urol. 2011;185(5):1583ā€“90.

    PubMedĀ  Google ScholarĀ 

  829. Assadipour Y, Sadowski SM, Alimchandani M, Quezado M, Steinberg SM, Nilubol N, Patel D, Prodanov T, Pacak K, Kebebew E. SDHB mutation status and tumor size but not tumor grade are important predictors of clinical outcome in pheochromocytoma and abdominal paraganglioma. Surgery. 2017;161(1):230ā€“9.

    PubMedĀ  Google ScholarĀ 

  830. Press D, Akyuz M, Dural C, Aliyev S, Monteiro R, Mino J, Mitchell J, Hamrahian A, Siperstein A, Berber E. Predictors of recurrence in pheochromocytoma. Surgery. 2014;156(6):1523ā€“7. discussion 1527-8.

    PubMedĀ  Google ScholarĀ 

  831. Kim KY, Kim JH, Hong AR, Seong MW, Lee KE, Kim SJ, Kim SW, Shin CS, Kim SY. Disentangling of malignancy from benign pheochromocytomas/paragangliomas. PLoS One. 2016;11(12):e0168413.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  832. Lucon AM, Pereira MA, MendonƧa BB, Halpern A, Wajchenbeg BL, Arap S. Pheochromocytoma: study of 50 cases. J Urol. 1997;157(4):1208ā€“12.

    PubMedĀ  CASĀ  Google ScholarĀ 

  833. Thompson LD. Pheochromocytoma of the Adrenal gland Scaled Score (PASS) to separate benign from malignant neoplasms: a clinicopathologic and immunophenotypic study of 100 cases. Am J Surg Pathol. 2002;26:551ā€“66.

    PubMedĀ  Google ScholarĀ 

  834. Lupşan N, Resiga L, Boşca AB, Georgiu C, Crişan D, Mirescu C, Constantin AM, Şimon I, Şovrea AS. Diagnostic reevaluation of 17 cases of pheochromocytomaĀ ā€“ a retrospective study. Romanian J Morphol Embryol. 2016;57(2 Suppl):651ā€“61.

    Google ScholarĀ 

  835. Loeffel SC, Gillespie GY, Mirmiran SA, Miller EW, Golden P, Askin FB, Siegal GP. Cellular immunolocalization of S100 protein within fixed tissue sections by monoclonal antibodies. Arch Pathol Lab Med. 1985;109(2):117ā€“22.

    PubMedĀ  CASĀ  Google ScholarĀ 

  836. Fishbein L, Nathanson KL. Pheochromocytoma and paraganglioma: understanding th e complexities of the genetic background. Cancer Genet. 2012;205(1ā€“2):1ā€“11.

    PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  837. Gimenez-Roqueplo AP, Burnichon N, Amar L, Favier J, Jeunemaitre X, Plouin PF. Recent advances in the genetics of phaeochromocytoma and functional paraganglioma. Clin Exp Pharmacol Physiol. 2008;35(4):376ā€“9.

    PubMedĀ  CASĀ  Google ScholarĀ 

  838. King KS, Pacak K. Familial pheochromocytomas and paragangliomas. Mol Cell Endocrinol. 2014;386(1ā€“2):92ā€“100.

    PubMedĀ  CASĀ  Google ScholarĀ 

  839. Grubbs EG, Rich TA, Ng C, Bhosale PR, Jimenez C, Evans DB, Lee JE, Perrier ND. Long-term outcomes of surgical treatment for hereditary pheochromocytoma. J Am Coll Surg. 2013;216(2):280ā€“9.

    PubMedĀ  Google ScholarĀ 

  840. Comstock JM, Willmore-Payne C, Holden JA, Coffin CM. Composite pheochromocytoma: a clinicopathologic and molecular comparison with ordinary pheochromocytoma and neuroblastoma. Am J Clin Pathol. 2009;132(1):69ā€“73.

    PubMedĀ  CASĀ  Google ScholarĀ 

  841. Choi YM, Sung TY, Kim WG, Lee JJ, Ryu JS, Kim TY, Kim WB, Hong SJ, Song DE, Shong YK. Clinical course and prognostic factors in patients with malignant pheochromocytoma and paraganglioma: a single institution experience. J Surg Oncol. 2015;112(8):815ā€“21.

    PubMedĀ  Google ScholarĀ 

  842. Ayala-Ramirez M, Feng L, Johnson MM, Ejaz S, Habra MA, Rich T, Busaidy N, Cote GJ, Perrier N, Phan A, Patel S, Waguespack S, Jimenez C. Clinical risk factors for malignancy and overall survival in patients with pheochromocytomas and sympathetic paragangliomas: primary tumor size and primary tumor location as prognostic indicators. J Clin Endocrinol Metab. 2011;96(3):717ā€“25.

    PubMedĀ  CASĀ  Google ScholarĀ 

  843. Whittle SB, Smith V, Doherty E, Zhao S, McCarty S, Zage PE. Overview and recent advances in the treatment of neuroblastoma. Expert Rev Anticancer Ther. 2017;17(4):369ā€“86.

    PubMedĀ  CASĀ  Google ScholarĀ 

  844. Vo KT, Matthay KK, Neuhaus J, London WB, Hero B, Ambros PF, Nakagawara A, Miniati D, Wheeler K, Pearson AD, Cohn SL, DuBois SG. Clinical, biologic, and prognostic differences on the basis of primary tumor site in neuroblastoma: a report from the international neuroblastoma risk group project. J Clin Oncol. 2014;32(28):3169ā€“76.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  845. Lonergan GJ, Schwab CM, Suarez ES, Carlson CL. Neuroblastoma, ganglioneuroblastoma, and ganglioneuroma: radiologic-pathologic correlation. Radiographics. 2002;22(4):911ā€“34. 42.

    PubMedĀ  Google ScholarĀ 

  846. Lin X, Wu D, Chen C, Zheng N. Clinical characteristics of adrenal tumors in children: a retrospective review of a 15-year single-center experience. Int Urol Nephrol. 2017;49(3):381ā€“5.

    PubMedĀ  Google ScholarĀ 

  847. He WG, Yan Y, Tang W, Cai R, Ren G. Clinical and biological features of neuroblastic tumors: a comparison of neuroblastoma and ganglioneuroblastoma. Oncotarget. 2017;8(23):37730ā€“9.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  848. Arakawa A, Oguma E, Aihara T, Kishimoto H, Kikuchi A, Hanada R, Koh K. Long-term follow-up results of the observation program for neuroblastoma detected at 6-month mass screening. J Pediatr. 2014;165(4):855ā€“7.e1.

    PubMedĀ  Google ScholarĀ 

  849. Irwin MS, Park JR. Neuroblastoma: paradigm for precision medicine. Pediatr Clin N Am. 2015;62(1):225ā€“56.

    Google ScholarĀ 

  850. Bielle F, FrĆ©neaux P, Jeanne-Pasquier C, Maran-Gonzalez A, Rousseau A, Lamant L, Paris R, Pierron G, Nicolas AV, Sastre-Garau X, Delattre O, Bourdeaut F, Peuchmaur M. PHOX2B immunolabeling: a novel tool for the diagnosis of undifferentiated neuroblastomas among childhood small round blue-cell tumors. Am J Surg Pathol. 2012;36(8):1141ā€“9.

    PubMedĀ  Google ScholarĀ 

  851. Duijkers FA, Gaal J, Meijerink JP, Admiraal P, Pieters R, de Krijger RR, van Noesel MM. High anaplastic lymphoma kinase immunohistochemical staining in neuroblastoma and ganglioneuroblastoma is an independent predictor of poor outcome. Am J Pathol. 2012;180(3):1223ā€“31.

    PubMedĀ  CASĀ  Google ScholarĀ 

  852. Dabbs DJ, Thompson L. Diagnostic immunohistochemistry: theranostic and genomic applications, 4e. Philadelphia: Elsevier Saunders; 2013. Print.

    Google ScholarĀ 

  853. Davis JL, Matsumura L, Weeks DA, Troxell ML. PAX2 expression in Wilms tumors and other childhood neoplasms. Am J Surg Pathol. 2011;35(8):1186ā€“94.

    PubMedĀ  Google ScholarĀ 

  854. Chan ES, Pawel BR, Corao DA, Venneti S, Russo P, Santi M, Sullivan LM. Immunohistochemical expression of glypican-3 in pediatric tumors: an analysis of 414 cases. Pediatr Dev Pathol. 2013;16(4):272ā€“7.

    PubMedĀ  Google ScholarĀ 

  855. Ambros PF, Ambros IM, Brodeur GM, Haber M, Khan J, Nakagawara A, Schleiermacher G, Speleman F, Spitz R, London WB, Cohn SL, Pearson AD, Maris JM. International consensus for neuroblastoma molecular diagnostics: report from the International Neuroblastoma Risk Group (INRG) Biology Committee. Br J Cancer. 2009;100(9):1471ā€“82.

    PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  856. Cohn SL, Pearson AD, London WB, Monclair T, Ambros PF, Brodeur GM, Faldum A, Hero B, Iehara T, Machin D, Mosseri V, Simon T, Garaventa A, Castel V, Matthay KK, INRG Task Force. The International Neuroblastoma Risk Group (INRG) classification system: an INRG Task Force report. J Clin Oncol. 2009;27(2):289ā€“97.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  857. Bautista DV, Asch M, Kovacs K, Murray D. Adrenal myelolipomatous nodules mimicking adrenal neoplasms: report of three cases. Can J Surg. 1989;32(1):51ā€“5.

    PubMedĀ  CASĀ  Google ScholarĀ 

  858. Goetz SP, Niemann TH, Robinson RA, Cohen MB. Hematopoietic elements associated with adrenal glands. A study of the spectrum of change in nine cases. Arch Pathol Lab Med. 1994;118(9):895ā€“6.

    PubMedĀ  CASĀ  Google ScholarĀ 

  859. Paulsen SD, Nghiem HV, Korobkin M, et al. Changing role of imaging-guided percutaneous biopsy of adrenal masses: evaluation of 50 adrenal biopsies. Am J Roentgenol. 2004;182:1033ā€“7.

    Google ScholarĀ 

  860. Barron SH, Emanuel B. Adrenal cyst. A case report and a review of the pediatric literature. J Pediatr. 1961;59:592ā€“9.

    PubMedĀ  CASĀ  Google ScholarĀ 

  861. Foster DG. Adrenal cysts. Review of literature and report of case. Arch Surg. 1966;92(1):131ā€“43.

    PubMedĀ  CASĀ  Google ScholarĀ 

  862. Sebastiano C, Zhao X, Deng FM, Das K. Cystic lesions of the adrenal gland: our experience over the last 20 years. Hum Pathol. 2013;44(9):1797ā€“803.

    PubMedĀ  Google ScholarĀ 

  863. Erickson LA, Lloyd RV, Hartman R, et al. Cystic adrenal neoplasms. Cancer. 2004;101:1537ā€“44.

    PubMedĀ  Google ScholarĀ 

  864. Chien HP, Chang YS, Hsu PS, Lin JD, Wu YC, Chang HL, Chuang CK, Tsuei KH, Hsueh C. Adrenal cystic lesions: a clinicopathological analysis of 25 cases with proposed histogenesis and review of the literature. Endocr Pathol. 2008;19(4):274ā€“81.

    PubMedĀ  Google ScholarĀ 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Priya Rao .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

Ā© 2020 Springer Nature Switzerland AG

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Rao, P. et al. (2020). Genitourinary Pathology (Including Adrenal Gland). In: Moran, C.A., Kalhor, N., Weissferdt, A. (eds) Oncological Surgical Pathology . Springer, Cham. https://doi.org/10.1007/978-3-319-96681-6_16

Download citation

  • DOI: https://doi.org/10.1007/978-3-319-96681-6_16

  • Published:

  • Publisher Name: Springer, Cham

  • Print ISBN: 978-3-319-96680-9

  • Online ISBN: 978-3-319-96681-6

  • eBook Packages: MedicineMedicine (R0)

Publish with us

Policies and ethics