Skip to main content

Pathology of Peritoneal Surface Malignancies

  • Chapter
Treatment of Peritoneal Surface Malignancies

Part of the book series: Updates in Surgery ((UPDATESSURG))

  • 909 Accesses

Abstract

The role of the pathologist in a team dealing with patients affected by PSM is crucial. The steps of this work are not merely diagnostic; in fact, before diagnosis is achieved, the pathologist must collect a great deal of information necessary for analyzing the individual case to enable the surgeon to interpret correctly the macroscopic pattern presented by the patient. The majority of patients undergoing surgical procedures for PSM have already been given systemic chemotherapy, and the pathological pattern at the time of the surgical intervention is affected by the grade of therapeutic response. In cases in which lesions are not obvious, it is important to detect and analyze all areas of minimal alteration, both in the serosal surface and in deeper locations (as in visceral specimens such as large- and small-bowel tracts, uterus and adnexa, bladder, gall bladder, and other such areas). It is even more important to apply this protocol in cases in which superficial macroscopic lesions have virtually disappeared. For correct staging, it must be determined with as much certainty as possible that no microscopic residue remains.

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 84.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Hardcover Book
USD 109.99
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. Nik NN, Vang R, Shih IM, Kurman RJ (2014) Origin and pathogenesis of pelvic (ovarian, tubal, and primary peritoneal) serous carcinoma. Ann Rev Pathol Mech Dis 9:27–45

    Article  CAS  Google Scholar 

  2. Shih I, Kurman RJ (2004) Ovarian tumorigenesis: a proposed model based on morphological andmolecular genetic analysis. Am J Pathol 164:1511–1518

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  3. Kurman RJ, Shih I (2011) Molecular pathogenesis and extraovarian origin of epithelial OC shifting the paradigm. Hum Pathol 42:918–931

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  4. Jones S, Wang TL, Shih I et al (2010) Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma. Science 330:228–231

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  5. Wiegand KC, Shah SP, Al-Agha OM et al (2010) ARID1A mutations in endometriosis associated ovarian carcinomas. N Engl J Med 363:1532–1543

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  6. Cho KR, Shih I (2009) Ovarian cancer. Annu Rev Pathol Mech Dis 4:287–313

    Article  CAS  Google Scholar 

  7. Piek JM, van Diest PJ, Zweemer RP et al (2001) Dysplastic changes in prophylactically removed fallopian tubes of women predisposed to developing OC. J Pathol 195:451–456

    Article  CAS  PubMed  Google Scholar 

  8. Medeiros F, Muto MG, Lee Y et al (2006) The tubal fimbria is a preferred site for early adenocarcinoma in women with familial OC syndrome. Am J Surg Pathol 30:230–236

    Article  PubMed  Google Scholar 

  9. Kindelberger DW, Lee Y, Miron A et al (2007) Intraepithelial carcinoma of the fimbria and pelvic serous carcinoma: evidence for a causal relationship. Am J Surg Pathol 31:161–169

    Article  PubMed  Google Scholar 

  10. Piek JM, Verheijen RH, Kenemans P et al (2003) BRCA1/2-related OCs are of tubal origin: a hypothesis. Gynecol Oncol 90:491

    Article  PubMed  Google Scholar 

  11. Kuhn E, Meeker A, Wang TL et al (2010) Shortened telomeres in serous tubal intraepithelial carcinoma, an early event in ovarian high-grade serous carcinogenesis. Am J Surg Pathol 34:829–836

    Article  PubMed  Google Scholar 

  12. Marquez RT, Baggerly KA, Patterson AP et al (2005) Patterns of gene expression in different histiotypes of epithelial OC correlate with those in normal fallopian tube, endometrium, and colon. Clin Cancer Res 11:6116–126

    Article  CAS  PubMed  Google Scholar 

  13. Di Giorgio A, Cardi M, Biacchi D et al (2013) Depth of colorectal-wall invasion and lymphnode involvement as major outcome factors influencing surgical strategy in patients with advanced and recurrent OC with diffuse peritoneal metastases. World J Surg Oncol 11:64–72

    Article  PubMed Central  PubMed  Google Scholar 

  14. Scarabelli C, Gallo A, Franceschi S et al (2000) Primary cytoreductive surgery with rectosigmoid colon resection for patients with advanced epithelial ovarian carcinoma. Cancer 88:389–397

    Article  CAS  PubMed  Google Scholar 

  15. Park JY, Seo SS, Kang S et al (2006) The benefits of low anterior en bloc resection as part of cytoreductive surgery for advanced primary and recurrent epithelial OC patients outweigh morbidity concerns. Gynecol Oncol 103:977–984

    Article  PubMed  Google Scholar 

  16. Dvoretsky PM, Richards KA, Angel C et al (1988) Distribution of disease at autopsy in 100 women with OC. Hum Pathol 19:57–63

    Article  CAS  PubMed  Google Scholar 

  17. Salani R, Diaz-Montes T, Giuntoli RL, Bristow RE (2007) Surgical management of mesenteric lymph node metastasis in patients undergoing rectosigmoid colectomy for locally advanced ovarian carcinoma. Ann Surg Oncol 14:3552–3557

    Article  PubMed  Google Scholar 

  18. Baiocchi G, Cestari LA, Macedo MP et al (2011) Surgical implications of mesenteric lymph node metastasis from advanced OC after bowel resection. J Surg Oncol 104:250–254

    Article  PubMed  Google Scholar 

  19. Baratti D, Kusamura S, Nonaka D et al (2008) Pseudomyxoma Peritonei: Clinical Pathological and Biological Prognostic Factors in Patients Treated with Cytoreductive Surgery and Hyperthermic Intraperitoneal Chemotherapy (HIPEC). Ann Surg Oncol 15:526–534

    Article  PubMed  Google Scholar 

  20. Szych C, Staebler A, Connolly DC et al (1999) Molecular genetic evidence supporting the clonality and appendiceal origin of pseudomyxoma peritonei in women. Am J Pathol 154:1849–1855

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  21. Carr NJ, Emory TS, Sobin LH (2002) Epithelial neoplasms of the appendix and colorectum: an analysis of cell proliferation, apoptosis and expression of p53, CD44, bcl-2. Arch Pathol Lab Med 126:837–841

    CAS  PubMed  Google Scholar 

  22. Ronnett BM, Yan H, Kurman RJ et al (2001) Patients with pseudomyxoma peritonei associated with disseminated peritoneal adenomucinosis have a significantly more favorable prognosis than patients with peritoneal mucinous carcinomatosis. Cancer 92:85–91

    Article  CAS  PubMed  Google Scholar 

  23. Bradley RF, Stewart JH, Russell GB et al (2006) Pseudomyxoma peritonei of appendiceal origin: a clinicopathological analysis of 101 patients uniformly treated at a single institution, with literature review. Am J Surg Pathol 30:551–559

    Article  PubMed  Google Scholar 

  24. Maehara Y, Hasuda S, Koga TY (2000) Postoperative outcome and sites of recurrence in patients following curative resection of GIC. Br J Surg 87:353–357

    Article  CAS  PubMed  Google Scholar 

  25. Thomassen I, van Gestel YR, van Ramshorst B et al (2014) Peritoneal carcinomatosis of gastric origin: A population-based study on incidence, survival and risk factors. Int J Cancer 134:622–628

    Article  CAS  PubMed  Google Scholar 

  26. Kodera Y (2013) Gastric cancer with minimal peritoneal metastasis: is this a sign to give up or to treat more aggressively? Nagoya J Med Sci 75:3–10

    PubMed  Google Scholar 

  27. Delotte J, Desantis M, Frigenza M et al (2014) Cytoreductive surgery with hyperthermic intraperitoneal chemotherapy for the treatment of endometrial cancer with peritoneal carcinomatosis. Eur J Obstet Gynecol Reprod Biol 172:11–114

    Article  Google Scholar 

  28. Bakrin N, Cotte E, Sayag-Beaujard A et al (2010) Cytoreductive surgery with hyperthermic intraperitoneal chemotherapy for the treatment of recurrent endometrial carcinoma confined to the peritoneal cavity. Int J Gynecol Cancer 20:809–814

    Article  PubMed  Google Scholar 

  29. Santeufemia DA, Lumachi F, Basso SM (2013) Cytoreductive surgery with hyperthermic intraperitoneal chemotherapy as salvage treatment for a late wound recurrence of endometrial cancer. Anticancer Res 33:1041–1044

    PubMed  Google Scholar 

  30. Mc Lemore EC, Pockaj BA, Reynolds C et al (2005) Breast cancer: presentation and intervention in women with gastrointestinal metastases and carcinomatosis. Ann Surg Oncol 12:886–894

    Article  Google Scholar 

  31. Tuthill M, Pell R, Giuliani R et al (2009) Peritoneal disease in breast cancer: a specific entity with an extremely poor prognosis. Eur J Cancer 45:2146–2149

    Article  PubMed  Google Scholar 

  32. Cardi M, Sammartino P, Framarino ML et al (2013) Treatment of peritoneal carcinomatosis from breast cancer by maximal cytoreduction and HPEC: A preliminary report on 5 cases. The Breast 22:845–849

    Article  PubMed  Google Scholar 

  33. Thomassen I, Lemmens VEPP, Nienhuijs SW et al (2013) Incidence, Prognosis, and Possible Treatment Strategies of Peritoneal Carcinomatosis of Pancreatic Origin. Pancreas 42:72–75

    Article  PubMed  Google Scholar 

  34. Blastik M, Plavecz E, Zalatnai A (2011) Pancreatic carcinomas in a 60-year, institute-based autopsy material with special emphasis of metastatic pattern. Pancreas 40:478–480

    Article  PubMed  Google Scholar 

  35. Morizane C, Okusaka T, Morita S et al (2011) Construction and validation of a prognostic index for patients with metastatic pancreatic adenocarcinoma. Pancreas 40:415–421

    Article  PubMed  Google Scholar 

  36. Shibata K, Matsumoto T, Yada K et al (2005) Factors Predicting Recurrence After Resection of Pancreatic Ductal Carcinoma. Pancreas 31:69–73

    Article  PubMed  Google Scholar 

  37. Farma JM, Pingpank JF, Libutti SK et al (2005) Limited Survival in Patients With Carcinomatosis From Foregut Malignancies After Cytoreduction and Continuous Hyperthermic Peritoneal Perfusion. J Gastrointest Surg 9:1346–1353

    Article  PubMed  Google Scholar 

  38. Kimura H, Fushida S, Mukawa A et al (2009) A resected case of effective treatment with S-1/gemcitabine and paclitaxel combination chemotherapy for advanced pancreatic cancer with peritoneal and liver metastases]. Gan To Kagaku Ryoho 36:1191–1194

    PubMed  Google Scholar 

  39. Vasseur B, Cadiot G, Zins M et al (1996) Peritoneal Carcinomatosis in Patients with Digestive Endocrine Tumors. Cancer 78:1686–1692

    Article  CAS  PubMed  Google Scholar 

  40. Raptopoulos V (1985) Peritoneal mesothelioma. Crit Rev Diagn Imaging 24:293–328

    CAS  PubMed  Google Scholar 

  41. De Pangher Manzini V (2005) Malignant peritoneal mesothelioma. Tumori 91:1–5

    PubMed  Google Scholar 

  42. Raju U, Fine G, Greenawald KA, Ohorodnik JM (1989) Primary papillary serous neoplasia of the peritoneum: a clinicopathologic and ultrastructural study of eight cases. Hum Pathol 20:426–436

    Article  CAS  PubMed  Google Scholar 

  43. Seidman JD, Zhao P, Yemelyanova A et al (2011) “Primary peritoneal” high-grade serous carcinoma is very likely metastatic from serous tubal intraepithelial carcinoma: assessing the new paradigm of ovarian and pelvic serous carcinogenesis and its implications for screening for OC. Gynecol Oncol 120:470–473

    Article  PubMed  Google Scholar 

  44. Schorge JO, Muto MG, Lee SJ et al (2000) BRCA1-related papillary serous carcinoma of the peritoneum has a unique molecular pathogenesis. Cancer Res 60:1361–1364

    CAS  PubMed  Google Scholar 

  45. Gerald WL, Ladanyi M, de Alava E et al (1998) Clinical, pathologic, and molecular spectrum of tumors associated with t(11;22)(p13;q12): desmoplastic small round-cell tumor and its variants. J Clin Oncol 16:3028–3036

    CAS  PubMed  Google Scholar 

  46. Schwarz RE, Gerald WL, Kushner BH et al (1998) Desmoplastic small round cell tumors: prognostic indicators and results of surgical management. Ann Surg Oncol 5:416–422

    Article  CAS  PubMed  Google Scholar 

  47. Lal DR, Su WT, Wolden SL et al (2005) Results of multimodal treatment for desmoplastic small round cell tumors. J Pediatr Surg 40:251–255

    Article  PubMed  Google Scholar 

  48. Hassan I, Shyyan R, Donohue JH et al (2005) Intraabdominal desmoplastic small round cell tumors: a diagnostic and therapeutic challenge. Cancer 104:1264–1270

    Article  PubMed  Google Scholar 

  49. Subbiah V, Viny AD, Anderson PM et al (2012) Optimizing the therapy of desmoplastic small round cell tumor: combined experience from the two major cancer centers. J Clin Oncol 30:10021

    Article  Google Scholar 

  50. Hayes-Jordan A, Anderson P, Curley S et al (2007) Continuous hyperthermic peritoneal perfusion for desmoplastic small round cell tumor. J Pediatr Surg 42:E29–E32

    Article  PubMed  Google Scholar 

  51. Hayes-Jordan A, Green H, Fitzgerald N et al (2010) Novel treatment for desmoplastic small round cell tumor: hyperthermic intraperitoneal perfusion. J Pediatr Surg 45:1000–1006

    Article  PubMed  Google Scholar 

  52. Chang F (2006) Desmoplastic small round cell tumors: cytologic, histologic, and immuno-histo-chemical features. Arch Pathol Lab Med 130:728–732

    PubMed  Google Scholar 

  53. Bilimoria MM, Holtz DJ, Mirza NQ et al (2002) Tumor volume as a prognostic factor for sarcomatosis. Cancer 94:2441–2446

    Article  PubMed  Google Scholar 

  54. National Cancer Institute, U.S. National Institutes of Health (2009) Surveillance epidemiology and end results. Available at http/www.seer.cancer.gov. Accessed May 31, 2014

    Google Scholar 

  55. Munene G, Mack LA, Temple WJ (2011) Systematic review on the efficacy of multimodal treatment of sarcomatosis with cytoreduction and intraperitoneal chemotherapy. Ann Surg Oncol 18:207–213

    Article  PubMed  Google Scholar 

  56. Berthet B, Sugarbaker TA, Chang D et al (1999) Quantitative methodologies for selection of patients with recurrent abdominopelvic sarcoma for treatment. Eur J Cancer 35:413–419

    Article  CAS  PubMed  Google Scholar 

  57. Rossi CR, Deraco M, De Simone M et al (2004) Hyperthermic intraperitoneal intraoperative chemotherapy after cytoreductive surgery for the treatment of abdominal sarcomatosis: Clinical outcome and prognostic factors in 60 consecutive patients. Cancer 100:1943–1950

    Article  PubMed  Google Scholar 

  58. Baumgartner JM, Ahrendt SA, Pingpank JF et al (2013) Aggressive Locoregional Management of Recurrent Peritoneal Sarcomatosis. J Surg Oncol 107:329–334

    Article  CAS  PubMed  Google Scholar 

  59. Salti GI, Ailabouni L, Undevia D (2012) Cytoreductive Surgery and Hyperthermic Intraperitoneal Chemotherapy for the Treatment of Peritoneal Sarcomatosis. Ann Surg Oncol 19:1410–1415

    Article  PubMed  Google Scholar 

  60. Bonvalot S, Cavalcanti A, Le Pe choux C et al (2005) Randomized trial of cytoreduction followed by intraperitoneal chemotherapy versus cytoreduction alone in patients with peritoneal sarcomatosis. Eur J Surg Oncol 31:917–923

    Article  CAS  PubMed  Google Scholar 

  61. Rossi CR, Casali P, Kusamura S (2008) The Consensus Statement on the Locoregional Treatment of Abdominal Sarcomatosis. J Surg Oncol 98:291–294

    Article  PubMed  Google Scholar 

  62. Oei TN, Jagannathan JP, Ramaiya N, Ros PR (2010) Peritoneal Sarcomatosis Versus Peritoneal Carcinomatosis: Imaging Findings at MDCT. AJR 195:W229–W235

    Article  PubMed  Google Scholar 

  63. Licht JD, Weissmann LB, Antman K (1988) Gastrointestinal sarcomas. Semin Oncol 15:181–188

    CAS  PubMed  Google Scholar 

  64. Miettinen M, Virolainen M, Sarlomo-Rikala M (1995) Gastrointestinal stromal tumors - Value of CD34 antigen in their identification and separation from true leiomyomas and schwannomas. Am J Surg Pathol 19:207–216

    Article  CAS  PubMed  Google Scholar 

  65. Katz SC, DeMatteo RP (2008) Gastrointestinal stromal tumors and leiomyosarcoma. J Surg Oncol 97:350–359

    Article  PubMed  Google Scholar 

  66. Miettinen M, Monihan JM, Sarlomo- Rikala M et al (1999) Gastrointestinal stromal tumors/smooth muscle tumors (GISTs) primary to the omentum and mesentery. Am J Surg Pathol 23:1109–1118

    Article  CAS  PubMed  Google Scholar 

  67. Burkill GJ, Badran M, Thomas JM (2003) Malignant gastrointestinal stromal tumor: distribution, imaging features, and pattern of metastatic spread. Radiology 226:527–532

    Article  PubMed  Google Scholar 

  68. Kindblom LG, Remotti HE, Aldenborg F, Meis-Kindblom JM (1998) Gastrointestinal pacemaker cell tumor (GIPACT). Gastrointestinal stromal tumors show phenotypic characteristics of the interstitial cell of Cajal. Am J Pathol 152:1259–1269

    PubMed Central  CAS  PubMed  Google Scholar 

  69. Cheng EY, Springfield DS, Mankin HJ (1995) Frequent incidence of extrapulmonary sites of initial metastasis in patients with liposarcoma. Cancer 75:1120–1127

    Article  CAS  PubMed  Google Scholar 

  70. Pearlstone DB, Pisters PW, Bold RJ et al (1999) Patterns of recurrence in extremity liposarcoma: implications for staging and follow-up. Cancer 85:85–92

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Antonio Ciardi .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2015 Springer-Verlag Italia

About this chapter

Cite this chapter

Ciardi, A., Di Giorgio, A. (2015). Pathology of Peritoneal Surface Malignancies. In: Di Giorgio, A., Pinto, E. (eds) Treatment of Peritoneal Surface Malignancies. Updates in Surgery. Springer, Milano. https://doi.org/10.1007/978-88-470-5711-1_4

Download citation

  • DOI: https://doi.org/10.1007/978-88-470-5711-1_4

  • Publisher Name: Springer, Milano

  • Print ISBN: 978-88-470-5710-4

  • Online ISBN: 978-88-470-5711-1

  • eBook Packages: MedicineMedicine (R0)

Publish with us

Policies and ethics