Home Random Page


CATEGORIES:

BiologyChemistryConstructionCultureEcologyEconomyElectronicsFinanceGeographyHistoryInformaticsLawMathematicsMechanicsMedicineOtherPedagogyPhilosophyPhysicsPolicyPsychologySociologySportTourism






Sex Cord-Stromal Tumors

Leydig cell tumor

Sertoli cell tumor

Most tumors in this group originate from intratubular germ cell neoplasia (ITGCN).[74] [75] ITGCN is seen adjacent to all germ cell tumors in adults except for spermatocytic seminoma

and epidermoid and dermoid cysts. With rare exceptions, it is also not seen in pediatric tumors (teratomas, yolk sac tumors). ITCGN is encountered with a high frequency in the following

conditions, listed in order of increasing risk: cryptorchidism, prior germ cell tumors, strong family history of germ cell tumor, androgen insensitivity syndrome, and gonadal dysgenesis

syndrome. Untreated ITGCN progresses to invasive germ cell tumor in approximately 50% of cases over 5 years of follow-up. Thus its significance is similar to carcinoma in situ in other

organs. If ITGCN is identified, it is treated by low-dose radiotherapy, which destroys the germ cells yet maintains the androgen production of the Leydig cells.

Neoplastic germ cells may differentiate along gonadal lines to give rise to seminoma or transform into a totipotential cell population that gives rise to nonseminomatous tumors. Such

totipotential cells may remain largely undifferentiated to form embryonal carcinoma or may differentiate along extraembryonic lines to form yolk sac tumors or choriocarcinomas.

Teratoma results from differentiation of the embryonic carcinoma cells along the lines of all three germ cell layers. Some studies suggest that seminomas are not end-stage neoplasms.

Similar to embryonal carcinomas, seminomas may also act as precursors from which other forms of testicular germ cell tumors originate. This view is supported by the fact that cells that

form intratubular germ cell neoplasias (the presumed precursors of all types of germ cell tumors) share morphologic and molecular characteristics with tumor cells in seminomas. Despite

the fascination of pathologists with the heterogeneity of testicular tumors, from a clinical standpoint the most important distinction in germ cell tumors is between seminomas and

nonseminomatous tumors. As will be discussed later, this clinical distinction has important bearings on treatment and prognosis.

Pathogenesis

As with all neoplasms, little is known about the ultimate cause of germ cell tumors. Several predisposing influences, however, are important: (1) cryptorchidism, (2) testicular dysgenesis,

and (3) genetic factors, all of which may contribute to a common denominator: germ cell maldevelopment. Reference has already been made to the increased incidence of neoplasms in

undescended testes. In most large series of testicular tumors, approximately 10% are associated with cryptorchidism. The higher the location of the undescended testicle (intra-abdominal

versus inguinal), the greater is the risk of developing cancer.

Patients with disorders of testicular development (testicular dysgenesis), including testicular feminization and Klinefelter syndrome, harbor an increased risk of developing germ cell



tumors. The risk is highest in patients with testicular feminization. In cryptorchid and dysgenetic testes, foci of intratubular germ cell neoplasms can be detected at a high frequency before

the development of invasive tumors.

Genetic predisposition also seems to be important, although no well-defined pattern of inheritance has been identified. In support, striking racial differences in the incidence of testicular

tumors can be cited. Blacks in Africa have an extremely low incidence of these neoplasms, which is unaffected by migration to the United States. Familial clustering has been reported, and

according to one study, sibs of affected individuals have a tenfold higher risk of developing testicular cancer than does the general population.

As with all tumors, genomic changes are undoubtedly important in the pathogenesis of testicular cancers. An isochromosome of the short arm of chromosome 12, i(12p), is found in

virtually all germ cell tumors, regardless of their histologic type. In the approximately 10% of cases in which i(12p) is not detected, extra genetic material derived from 12p is found on

other chromosomes. Obviously, dosage of genes located on 12p is critical for the pathogenesis of germ cell tumors, and several candidate genes have been identified, including a novel

gene, called DAD-R, that prevents apoptosis.[76] [77] It is of interest that i(12p) is also noted in ovarian germ cell neoplasms, suggesting that the events leading to this alteration may be

critical to the molecular pathogenesis of all germ cell neoplasms.

With this background of pathogenesis, we can discuss the morphologic patterns of germ cell tumors, followed by the clinical features that are common to most germinal tumors.

Seminoma

Seminomas are the most common type of germinal tumor (50%) and the type most likely to produce a uniform population of cells. They almost never occur in infants; they peak in the

thirties. An identical tumor arises in the ovary, where it is called dysgerminoma ( Chapter 22 ).

Morphology.

If not otherwise specified, the term "seminoma" refers to "classic" or "typical" seminoma. Spermatocytic seminoma, despite its nosologic similarity, is actually a distinct tumor; it has been

segregated into a separate category and will be discussed later.

Seminomas produce bulky masses, sometimes 10 times the size of the normal testis. The typical seminoma has a homogeneous, gray-white, lobulated cut surface, usually devoid of

hemorrhage or necrosis ( Fig. 21-23 ). In more than half of cases, the entire testis is replaced. Generally, the tunica albuginea is not penetrated, but occasionally, extension to the

epididymis, spermatic cord, or scrotal sac occurs.

Microscopically, the typical seminoma presents sheets of uniform cells divided into poorly demarcated lobules by delicate septa of fibrous tissue ( Fig. 21-24A ). The classic seminoma

cell is large and round to polyhedral and has a distinct cell membrane; a clear or watery-appearing cytoplasm; and a large,

Figure 21-23Seminoma of the testis appears as a fairly well circumscribed, pale, fleshy, homogeneous mass.

Figure 21-24Seminoma. A, Low magnification shows clear seminoma cells divided into poorly demarcated lobules by delicate septa. B, Microscopic examination reveals large cells with

distinct cell borders, pale nuclei, prominent nucleoli, and a sparse lymphocytic infiltrate.

Figure 21-25Embryonal carcinoma. In contrast to the seminoma illustrated in Figure 21-23 , the embryonal carcinoma is a hemorrhagic mass.

Figure 21-26Embryonal carcinoma shows sheets of undifferentiated cells as well as primitive glandular differentiation. The nuclei are large and hyperchromatic.

Figure 21-27Choriocarcinoma shows clear cytotrophoblastic cells with central nuclei and syncytiotrophoblastic cells with multiple dark nuclei embedded in eosinophilic cytoplasm.

Hemorrhage and necrosis are prominent.

Figure 21-28Teratoma of the testis. The variegated cut surface with cysts reflects the multiplicity of tissue found histologically.

Figure 21-29Teratoma of the testis consisting of a disorganized collection of glands, cartilage, smooth muscle, and immature stroma.

Figure 21-30Adult prostate. The normal prostate contains several distinct regions, including a central zone (CZ), a peripheral zone (PZ), a transitional zone (TZ), and a periurethral zone.

Most carcinomas arise from the peripheral glands of the organ and may be palpable during digital examination of the rectum. Nodular hyperplasia, in contrast, arises from more centrally

situated glands and is more likely to produce urinary obstruction early on than is carcinoma.

Figure 21-31Benign prostate gland with basal cell and secretory cell layer.

Figure 21-32Simplified scheme of the pathogenesis of prostatic hyperplasia. The central role of the stromal cells in generating dihydrotestosterone should be noted.

Figure 21-33Nodular prostatic hyperplasia. A, Well-defined nodules of BPH compress the urethra into a slitlike lumen. B, A microscopic view of a whole mount of the prostate shows

nodules of hyperplastic glands on both sides of the urethra.

Figure 21-34Adenocarcinoma of the prostate. Carcinomatous tissue is seen on the posterior aspect (lower left). Note the solid whiter tissue of cancer in contrast to the spongy appearance

of the benign peripheral zone on the contralateral side.

Figure 21-36 A, Photomicrograph of a small focus of adenocarcinoma of the prostate demonstrating small glands crowded in between larger benign glands. B, Higher magnification shows

several small malignant glands with enlarged nuclei, prominent nucleoli, and dark cytoplasm, compared to the larger benign gland (top).

Figure 21-35Metastatic osteoblastic prostatic carcinoma within vertebral bodies.

Figure 21-37Carcinoma of the prostate showing perineural invasion by malignant glands. Compare to a benign gland (left).

Figure 21-38 A, Low-grade (Gleason score 1 + 1 = 2) prostate cancer consisting of back to back, uniformly sized malignant glands. Glands contain eosinophilic intraluminal prostatic

crystalloids, a feature that is more commonly seen in cancer than in benign glands and more frequently seen in lower grade than in higher grade prostate cancer. B, Needle biopsy of the

prostate with variably sized, more widely dispersed glands of moderately differentiated (Gleason score 3 + 3 = 6) adenocarcinoma. C, Poorly differentiated Gleason score (5 + 5 = 10)

adenocarcinoma composed of sheets of malignant cells.

References

1. Bostwick DG, Eble JN (eds): Urologic Surgical Pathology. St. Louis, Mosby, 1997.

2. Murphy WM (ed): Urologic Pathology, 2nd ed. Philadelphia, WB Saunders, 1997.

3. Kottra JJ, Dunnick NR: Retroperitoneal fibrosis. Radiol Clin North Am 34:1259, 1996.

4. Smeulders N, Woodhouse CRJ: Neoplasia in adult extrophy patients. BJU Int 87:623, 2001.

5. de Vries CR, Freiha FS: Hemorrhagic cystitis: a review. J Urol 143:1, 1990.

6. Nickel JC: Interstitial cystitis: etiology, diagnosis, and treatment. Can Fam Physician 46:2430, 2000.

7. Long JR Jr, Althausen AF: Malacoplakia: a 25-year experience with a review of the literature. J Urol 141:1328, 1989.

8. Young RH.: Papillary and polypoid cystitis: a report of eight cases. Am J Surg Pathol 12:542, 1988.

9. Corica FA, et al: Intestinal metaplasia is not a strong risk factor for bladder cancer: study of 53 cases with long-term follow-up. Urol 50:427, 1997.

10. Young RH, Scully RE: Nephrogenic adenoma. Am J Surg Pathol 10:268, 1986.

11. Ford TF, et al: Adenomatous metaplasia (nephrogenic adenoma) of urothelium: an analysis of 70 cases. Br J Urol 57:427, 1985.

12. American Cancer Society: Cancer Facts and Figures. Atlanta, GA: American Cancer Society, 2003.

13. Murphy WM, et al: Tumors of the kidney, bladder, and related urinary structures. In Atlas of Tumor Pathology, 3rd series, fascicle 11. Washington, DC, Armed Forces Institute of

Pathology, 1994.

14. Taylor DC, et al: Papillary urothelial hyperplasia: a precursor to papillary neoplasms. Am J Surg Pathol 20:1481, 1996.

15. Epstein JI, et al: The World Health Organization/International Society of Urological Pathology consensus classification of urothelial (transitional cell) neoplasms of the urinary bladder.

Am J Surg Path 22:1435, 1998.

16. Bergkvist A, et al: Classification of bladder tumours based on the cellular pattern. Acta Chir Scand 130:371, 1965.

17. Mostofi FK, et al: Histological typing of urinary bladder tumours. In International Classification of Tumors, Vol. 19. Geneva, World Health Organization, 1973.

18. Jordan AM, et al: Transitional cell neoplasms of the urinary bladder: can biologic potential be predicted from histologic grading? Cancer 60:2766, 1987.

19. Cheville JC, et al: Inverted urothelial papilloma: is ploidy, MIB-1 proliferative activity, or p53 protein accumulation predictive of urothelial carcinoma? Cancer 88:632, 2000.

20. Witjes JA, et al: The prognostic value of a primary inverted papilloma of the urinary tract. J Urol 158:1500, 1997.

21. Gilbert HA, et al: The natural history of papillary transitional cell carcinoma of the bladder and its treatment in an unselected population on the basis of histologic grading. J Urol

119:488, 1978.

22. Heney NM, et al: Superficial bladder cancer: progression and recurrence. J Urol 130:1083, 1983.

23. Melamed MR, et al: Natural history and clinical behavior of in situ carcinoma of the human urinary bladder. Cancer 17:1533, 1964.

24. Melicow MM, Hollowell JW: Intra-urothelial cancer-carcinoma in situ. Bowen's disease of the urinary system: Discussion of thirty cases. J Urol 68:763, 1952.

25. Farrow GM, et al: Clinical observations on 69 cases of in situ carcinoma of the urinary bladder. Cancer Res 32:2794, 1977.

26. Elliot GB, et al: "Denuding cystitis" and in situ urothelial carcinoma. Arch Pathol 96:91, 1973.

27. Drew PA, et al: The nested variant of transitional cell carcinoma: an aggressive neoplasm with innocuous histology. Mod Pathol 9:898, 1996.

28. Talbert ML, Young RH: Carcinomas of the urinary bladder with deceptively benign-appearing foci. Am J Surg Pathol 13:374, 1989.

29. Amin MB, et al: Lymphoepithelioma-like carcinoma of the urinary bladder. Am J Surg Pathol 18:466, 1994.

30. Sakamoto N, et al: Urinary bladder carcinoma with neoplastic squamous component: a mapping study of 31 cases. Histopathology 21:135, 1992.

31. El-Bolkainy MN, et al: The impact of schistosomiasis on the pathology of bladder carcinoma. Cancer 48:2643, 1981.

32. Grignon DJ, et al: Primary adenocarcinoma of the urinary bladder: a clinicopathologic analysis of 72 cases. Cancer 67:2165, 1991.

33. Xiaoxu L, et al: Bladder adenocarcinoma: 31 reported cases. Can J Urol 8:1380, 2001.

34. Brandau S, Böhle A: Bladder: Molecular and genetic basis of carcinogenesis. Eur Urol 39:491, 2001.

35. Jung I, Messing E: Molecular mechanisms and pathways in bladder cancer development and progression. Cancer Control 7:325, 2000.

36. Cordon-Cardo C, et al: P53 mutations in human bladder cancer: genotypic versus phenotypic patterns. Cancer 56:347, 1994.

37. Gibas C, Gibas L: Cytogenetics of bladder cancer. Cancer Genet Cytogenet 95:108, 1997.

38. Cairns P, Sidransky D: Bladder cancer. In Vogelstein B, Kinzler A (eds): Genetic Basis of Human Cancer. New York, McGraw-Hill, 1998, pp 639–645.

39. Spruck CH, et al: Two molecular pathways for transitional carcinoma of the bladder. Cancer Res 54:784, 1994.

40. Holmang S, et al: Stage progression in Ta papillary urothelial tumors: relationship to grade, immunohistochemical expression of tumor markers, mitotic frequency and DNA ploidy. J

Urol 165:1124, 2001.

41. Malmström P-U, et al: Recurrence, progression, and survival in bladder cancer: a retrospective analysis of 232 patients with 5-year follow-up. Scand J Urol Nephrol 21:185, 1987.

42. Smith G, et al: Prognostic significance of biopsy results of normal-looking mucosa in cases of superficial bladder cancer. Br J Urol 55:665, 1983.

43. Melicow MM: Histological study of vesical urothelium intervening between gross neoplasms in total cystectomy. J Urol 68:261, 1952.

44. Murphy WM, Soloway MS. Developing carcinoma (dysplasia) of the urinary bladder. Pathol Annu 17:197, 1982.

45. Koss LG. Mapping of the urinary bladder: its impact on the concepts of bladder cancer. Hum Pathol 10:533, 1979.

46. Murphy WM, et al: Urinary cytology and bladder cancer: the cellular features of transitional cell neoplasms. Cancer 53:1555, 1984.

47. Murphy WM: Current status of urinary cytology in the evaluation of bladder neoplasms. Hum Pathol 21:886, 1990.

48. Koss LG: Diagnostic Cytology and Its Histopathologic Bases, 4th ed. Philadelphia, Lippincott Raven, 1992, p 890.

49. Koss LG: Diagnostic cytology of the urinary tract with histopathologic with histopathologic and clinical correlations. Philadelphia, Lippincott, 1995.

50. Herr HW, et al: Bacillus Calmette-Guérin therapy for special bladder cancer: a 10-year followup. J Urol 147:1020, 1992.

51. Lopez-Beltran A, et al: Carcinosarcoma and sarcomatoid carcinoma of the bladder: clinicopathological study of 41 cases. J Urol 159:1497, 1998.

52. Jones EC, et al: Inflammatory pseudotumor of the urinary bladder. Am J Surg Pathol 17:264, 1993.

53. Kempton CL, et al: Malignant lymphoma of the bladder: evidence from 36 cases that low-grade lymphoma of the MALT-type is the most common primary bladder lymphoma. Am J

Surg Pathol 21:1324, 1997.

54. Diamond DA, Ransley PG: Male epispadias. J Urol 154:2150, 1995.

55. Belman AB: Hypospadias update. Urology 49:166, 1997.

56. Davenport M: ABC of general surgery in children: problems with penis and prepuce. BMJ 312:299, 1996.

57. Edwards S: Balanitis and balanoposthitis: a review. Genitourin Med 72:155, 1996.

58. Cupp MR, et al: The detection of human papilloma virus deoxyribonucleic acid in intraepithelial, in situ, verrucous and invasive carcinoma of the penis. J Urol 154:1024, 1995.

59. Dillner J, et al: Etiology of squamous cell carcinoma of the penis. Scand J Urol Nephrol Suppl 205:189, 2000.

60. Cubilla AL, et al: Histologic classification of penile carcinoma and its relation to outcome in 61 patients with primary resection. Int J Surg Pathol 9:111, 2001.

61. Cubilla AL, et al: Morphological features of epithelial abnormalities and precancerous lesions of the penis. Scand J Urol Nephrol Suppl 205:215, 2000.

62. Burgers JK, et al: Penile cancer: clinical presentation, diagnosis, and staging. Urol Clin North Am 19:267, 1992.

63. Rozanski TA, Bloom D: The undescended testis: theory and management. Urol Clin North Am 22:107, 1995.

64. Hutson J, et al: Normal testicular descent and the etiology of cryptorchidism. Adv Anal Embryol Cell Biol 132:1, 1996.

65. Swerdlow AJ, et al: Risk of testicular cancer in cohort of boys with cryptorchidism. BMJ 314:1507, 1997.

66. Davenport M: ABC of general pediatric surgery: inguinal hernia, hydrocele, and the undescended testis. BMJ 312:564, 1996.

67. Forman D, et al: Aetiology of testicular cancer: association with congenital abnormalities, age at puberty, infertility and exercise. BMJ 308:1393, 1994.

68. Beutow SA: Epidemiology of testicular cancer. Epidemiol Rev 17:433, 1995.

69. Nistal M, Paniagua R: Testicular biopsy: contemporary interpretation. Urol Clin N Am 26:555, 1999.

70. Ulbright TM, et al: Tumors of the testis, adnexa, spermatic cord, and scrotum. In Atlas of Tumor Pathology, 3rd series, fascicle 25. Washington, DC: Armed Forces Institute of

Pathology, 1999.

71. Ulbright TM: Testis risk and prognostic factors: the pathologist's perspective. Urol Clin North Am 26:611, 1999.

72. Ulbright TM: Germ cell neoplasms of the testis. Am J Surg Pathol 17:1075, 1993.

73. Bosl GJ, Motzer RJ: Testicular germ-cell cancer. N Engl J Med 337:242, 1997.

74. Rorth M, et al: Carcinoma in situ in the testis. Scand J Urol Nephrol Suppl 205:166, 2000.

75. Looijenga LH, Oosterhuis JW: Pathogenesis of testicular germ cell tumours. Rev Reprod 4:90, 1999.

76. Looijenga LH, et al: Role of gain of 12p in germ cell tumor development. APMIS 111:161, 2003.

77. Rodriguez S, et al: Expression profile of genes from 12p in testicular germ cell tumors of adolescents and adults associated with I(12p) and amplification of 12p11.2-p12.1. Oncogene

22:1880, 2003.

78. Eble JN: Spermatocytic seminoma. Hum Pathol 25:1035, 1994.

79. Motzer RJ, et al: Teratoma with malignant transformation: diverse malignant histologies arising in men with germ cell tumors. J Urol 159:133, 1998.

80. Doherty AP, et al: The role of tumor markers in the diagnosis and treatment of testicular germ cell cancers. Br J Urol 79:247, 1997.

81. Leendert HJL, Oosterhuis JW: Clinical value of the x chromosome in testicular germ cell tumors. Lancet 363:6, 2004.

82. Dilworth JP, et al: Non-germ cell tumors of testis. Urology 37:399, 1991.

83. Kim I, et al: Leydig cell tumors of the testis: a clinicopathological analysis of 40 cases and review of the literature. Am J Surg Pathol 9:177, 1985.

84. Cheville JC, et al: Leydig cell tumor of the testis: a clinicopathologic, DNA content, and MIB-1 comparison of nonmetastasizing and metastasizing tumors. Am J Surg Pathol 22:1361,

1998.

85. Young RH, et al: Sertoli cell tumors of the testis, not otherwise specified: a clinicopathologic analysis of 60 cases. Am J Surg Pathol 22:709, 1998.

86. Ferry, JA, et al: Malignant lymphoma of the testis, epididymis, and spermatic cord: a clinicopathologic study of 69 cases with immunophenotypic analysis. Am J Surg Pathol 18:376,

1994.

87. McNeal JE: Normal and pathologic anatomy of prostate. Urology 17 (suppl):11, 1981.

88. Nickel JC: Prostatitis: Evolving management strategies. Urol Clin North Am 26:737, 1999.

89. Lipsky BA: Prostatitis and urinary tract infection in men: what's new; what's true? Am J Med 106:327, 1999.

90. Wise GJ, Silver DA: Fungal infections of the genitourinary system. J Urol 149:1377, 1993.

91. Foster CS: Pathology of benign prostatic hyperplasia. Prostate 9 (suppl):4, 2000.

92. Ramsey EW: Benign prostatic hyperplasia: a review. Can J Urol 7:1135, 2000.

93. Wong YC, Wang YZ: Growth factors and epithelial-stromal interactions in prostate cancer development. Int Rev Cytol 199:65, 2000.

94. McConnell JD, et al: The long-term effect of Doxazosin, Finasteride, and combination therapy on clinical progression of benign prostatic hyperplasia. New Engl J Med 349:2387, 2003.

95. Droller MJ: Medical approaches to the management of prostate disease. Br J Urol 79:42, 1997.

96. Walsh PC: Treatment of benign prostatic hyperplasia. N Engl J Med 335:586, 1996.

97. American Cancer Society: Cancer Facts and Figures. Atlanta, GA: American Cancer Society, 2004.

98. Ekman P: Genetic and environmental factors in prostate cancer genesis: identifying high-risk cohorts. Eur Urol 35:362, 1999.

99. Gronberg H: Prostate cancer epidemiology. Lancet 361:859, 2003.

100. Nelson WG, DeMarzo AM, Isaccs WB: Prostate cancer. New Engl J Med 349:366, 2003.

101. Yip I, et al: Nutrition and prostate cancer. Urol Clin North Am 26:403, 1999.

102. Boyle P, Severi G, and Giles GG: The epidemiology of prostate cancer. Urol Clin North Am 30:209, 2003.

103. Isaacs JT: Molecular markers of prostate cancer metastases. Am J Pathol 150:1511, 1997.

104. Ruijter E, et al: Molecular changes associated with prostate cancer development. Anal Quant Cytol Histol 23:67, 2001.

105. Primo NR Jr, et al: Molecular biology of prostate carcinogenesis. Crit Rev Oncol Hematol 32:197, 1999.

106. Jussi PE, Visakorpi T: Molecular genetics of prostate cancer. Ann Med 33:130, 2001.

107. Rhodes DR, et al: Multiplex biomarker approach for determining risk of prostate-specific antigen-defined recurrence of prostate cancer. J Natl Cancer Inst 95:661, 2003.

108. DeMarzo AM, et al: Pathologic and molecular aspects of prostate cancer. Lancet 361:955, 2003.

109. Byar DP, et al: Carcinoma of the prostate: prognostic evaluation of certain pathologic features in 208 radical prostatectomies. Cancer 30:5, 1972.

110. Potter SR, et al: Seminal vesicle invasion by prostate cancer: prognostic significance and therapeutic implications. Rev Urol 2:190, 2000.

111. Saitoh H, et al: Metastatic patterns of prostatic cancer: correlation between sites and number of organs involved. Cancer 54:3078, 1984.

112. Epstein JI: Diagnostic criteria of limited adenocarcinoma of the prostate on needle biopsy. Hum Pathol 26:223, 1995.

113. Epstein JI: Interpretation of Prostate Biopsies, 3rd ed. Philadelphia, Lippincott Williams & Wilkins, 2002.

114. Young RH, et al: Tumors of the prostate gland, seminal vesicles, male urethra, and penis. In Atlas of Tumor Pathology, 3rd series, fascicle 28. Washington, DC: Armed Forces

Institute of Pathology, 2000.

115. Rubin MA, et al: a-Methylacyl coenzyme A racemase as a tissue biomarker for prostate cancer. JAMA 287:1662–1670, 2002.

116. Haggman MJ, et al: The relationship between prostatic intrapeithelial neoplasia and prostate cancer: critical issues. J Urol 158:12, 1997.

117. McNeal JE, Bostwick DG. Intraductal dysplasia: a pre-malignant lesion of the prostate. Human Pathol 17:64, 1986.

118. Kronz JD, et al: Predicting cancer following a diagnosis of high grade prostatic intraepithelial neoplasia on needle biopsy: data on men with more than one follow-up biopsy. Am J

Surg Pathol 25:1079, 2001.

119. McNeal JE. Origin and development of carcinoma in the prostate. Cancer 23:24, 1969.

120. Epstein JI: Pathological assessment of the surgical specimen. Urol Clin North Am 28:567, 2001.

121. Gleason DF, et al: Prediction of prognosis for prostatic adenocarcinoma by combined histologic grading and clinical staging. J Urol 111:58, 1974.

122. Epstein JI, et al: The pathologic interpretation and significance of prostate biopsy findings: implications and current controversies. J Urol 166:402, 2001.

123. McNeal JE, et al: Histologic differentiation, cancer volume, and pelvic lymph node metastasis in adenocarcinoma of the prostate. Cancer 66:1225, 1990.

124. Epstein JI, et al: Pathological and clinical findings to predict tumor extent of non-palpable (stage T1c) prostate cancer. JAMA 271:368, 1994.

125. Matzkin H, et al: Stage T1a carcinoma of the prostate. Urology 43:11–21, 1994.

126. Sokoll LJ, Chan DW: Prostate-specific antigen: its discovery and biochemical characteristics. Urol Clin North Am 24:253, 1997.

127. Gretzer MD, Partin AW: PSA markers in prostate cancer. Urol Clin N Am 30:677, 2003.

128. Arcangeli CG, et al: Prostate-specific antigen as a screening test for prostate cancer: the United States experience. Urol Clin North Am 24:299, 1997.

129. Catalona WJ: Clinical utility of measurements of free and total prostate-specific antigen (PSA): a review. Prostate 7 (suppl):64, 1996.

130. Vashi AR, Oesterling JE: Percent free prostate-specific antigen: entering a new era in the detection of prostate cancer. Mayo Clin Proc 72:337, 1997.

131. Catalona WJ, et al: Use of the percentage of free prostate-specific antigen to enhance differentiation of prostate cancer from benign prostatic disease: a prospective multicenter clinical

trial. JAMA 279:1542, 1998.

132. Epstein JI: PSAP and PSA as immunohistochemical markers. Urol Clin North Am 20:757, 1993.

133. Armas OA, et al: Clinical and pathobiological effects of neoadjuvant total androgen ablation therapy on clinically localized prostatic adenocarcinoma. Am J Surg Pathol 18:979, 1994.

134. Bostwick DG, et al: Radiation injury of the normal and neoplastic prostate. Am J Surg Pathol 6:501, 1982.

135. Brinker DA, et al. Ductal adenocarcinoma of the prostate diagnosed on needle biopsy: correlation with clinical and radical prostatectomy findings and progression. Am J Surg Pathol

23:1471, 1999.

136. Little NA, et al: Squamous cell carcinoma of the prostate: 2 cases of a rare malignancy and review of the literature. J Urol 149:137, 1993.

137. Ro JY, et al: Mucinous adenocarcinoma of the prostate: histochemical and immunohistochemical studies. Hum Pathol 21:593, 1990.

138. Tetu B, et al: Small cell carcinoma of prostate. Part 1: a clinicopathologic study of 20 cases. Cancer 59:1803, 1987.

139. Wood DP, et al: Transitional cell carcinoma of the prostate in cystoprostatectomy specimens removed for bladder cancer. J Urol 141:346, 1989.

140. Oliai BR, et al: A clinicopathologic analysis of urothelial carcinomas diagnosed on prostate needle biopsy. Am J Surg Pathol 25:794, 2001.

141. Esrig D, et al: Transitional cell carcinoma involving the prostate with a proposed staging classification for stromal invasion. J Urol 156:1071, 1996.

142. Proppe KH, et al: Postoperative spindle cell nodules of genitourinary tract resembling sarcoma. Am J Surg Pathol 8:101, 1984.

143. Sahin AA, et al: Pseudosarcomatous fibromyxoid tumor of the prostate: a case report with immunohistochemical, electron microscopic, and DNA flow cytometric analysis. Am J Clin

Pathol 96:253, 1991.

144. Cheville JC, et al: Leiomyosarcoma of the prostate: report of 23 cases. Cancer 76:1422, 1995.

145. Gaudin PB, et al: Sarcomas and related proliferative lesions of specialized prostatic stroma. Am J Surg Pathol 22:148, 1998.

146. Lauwers GY, et al: Carcinosarcoma of the prostate. Am J Surg Pathol 17:342, 1993.

 


Date: 2016-04-22; view: 1987


<== previous page | next page ==>
Miscellaneous Anomalies. | Location and Manifestations of Infection
doclecture.net - lectures - 2014-2024 year. Copyright infringement or personal data (0.019 sec.)