Ovary: Epithelial tumors

2003-12-01   Lisa Lee-Jones 

1.Tumour Molecular Genetics Group, Institute of Medical Genetics, University of Wales College of Medicine, Heath Park, Cardiff, CF14 4XN, UK

Summary

Note

Ovarian epithelial tumours are thought to arise from the simple cuboidal surface epithelium of the ovary, and account for 75% of all ovarian tumours, and 90-95% of ovarian malignancies.

Classification

Note

Ovarian epithelial tumours are classified according to the following histological subtypes:
  • serous,
  • mucinous,
  • endometrioid,
  • clear cell,
  • Brenner,
  • transitional cell,
  • small cell,
  • mixed mesodermal and
  • undifferentiated.
Usually each subtype can be classified as benign, borderline (low malignant potential, LMP), or malignant (invasive).

Classification

Serous tumours are further subdivided into the following:
  • Serous cystadenoma
  • Borderline serous tumour
  • Serous cystadenocarcinoma
  • Adenofibroma
  • Cystadenofibroma

Mucinous tumours are further classified as:
  • Mucinous cystadenoma
  • Borderline mucinous tumour
  • Mucinous cystadenocarcinoma
  • Adenofibroma

Clinics and Pathology

Etiology

Epidemiology studies have provided data showing increased risk for ovarian cancer with greater numbers of ovulation cycles. Multiple pregnancies and use of oral contraceptives are thought to have a protective effect because of decreased ovulation and hormonal influences. There are two theories explaining for the association of decreased risk with decreased number of ovulation cycles:
1 Theory of incessant ovulation: repeated ovarian follicular rupture and subsequent repair results in increased likelihood of genetic alterations within the surface epithelium.
2. The Gonadotrophin Theory hypothesis: persistent stimulation of the ovaries by gonadotrophins, together with local effects of endogenous hormones, results in increased proliferation and mitotic activity of the surface epithelium. This is consistent with ovarian cancer being associated with high gonadotrophin states such as the menopause, and less commonly associated with low gonadotrophin states such as oral contraceptive use and high parity.

Epithelial ovarian carcinoma develops sporadically in about 90-95% of patients. Environmental and dietary factors are thought to have a role. These include use of talc on the perineum and vulva, asbestos, pelvic irradiation, viruses (particularly mumps), high-fat diet, and lactose consumption. Other factors are associated with an increased number of ovulation cycles: low parity, delayed childbearing, early menarche and late menopause. However, genetic factors are the most important risk factor for ovarian epithelial carcinoma (See Genetics section of this review for further details).

Factors that decrease the risk for ovarian cancer predominantly reduce the number of ovulation cycles a women encounters-such as the use of oral contraceptives, breast-feeding and multiparity. Long-term use of oral contraceptives has reduced the risk of ovarian cancer by more than 50% in unselected women. Decreased risk of ovarian cancer has also been associated with tubal ligation and hysterectomy.

Epidemiology

Epithelial ovarian cancer is the sixth most common cancer in women and is the second most common female genital tract malignancy after endometrial cancer. They are usually found in postmenopausal women and are the commonest cause of death among women with gynaecologic malignancies in the USA, accounting for approximately 15,000 deaths annually. The annual lifetime risk for ovarian cancer is 1.4 per 100 women in the USA. Epithelial ovarian cancer can occur in females as young as 15, however the mean presentation age is 56 years. The age-specific incidence gradually rises and peaks at 70 years of age (55 per 100,000 Caucasians), whereas it affects only 3 women per 100,000 before 30 years of age. The median age for ovarian adenocarcinoma (which accounts for 85-90% of all malignant tumours) is between 60-65 years. The LMP ovarian tumours present at a younger age; the mean age of diagnosis is 48 years, and no large peak of incidence is observed. Brenner tumours are diagnosed in peri- or postmenopausal women. The incidence of ovarian epithelial tumours varies globally, with highest rates being observed in Scandinavia, Israel and North America, whereas the lowest rates are found in developing countries and Japan. A racial predisposition to ovarian epithelial tumours is apparent, with lower risks for black women. Clear cell adenocarcinoma is more prevalent in Japanese than western countries.

Clinics

Most early ovarian carcinomas and the serous and mucinous cystadenomas are asymptomatic. Two-thirds of patients present with extensive intra-abdominal metastases. Patients with advanced carcinomas usually present with vague abdominal swelling or discomfort, abdominal bloating, dyspepsia and early satiety, lack of appetite, malaise, urinary frequency and weight change (either gain or loss). Pelvic examination revealing firmness, fixation, nodularity, lack of tenderness, ascites, or cul-de-sac nodules are indicative of malignancy. 50% of all ovarian carcinomas are bilateral. Malignant serous tumours constitute over 40% of invasive epithelial carcinomas. Both borderline and malignant serous tumours are often bilateral. Mucinous carcinomas are diagnosed at stage I in approximately half of patients, whereas serous tumours are usually diagnosed at advanced stages.

Brenner tumours are virtually always benign, and the exceptional malignant cases resemble transitional cell carcinoma of the bladder. As with the other types of ovarian neoplasm, it is usually asymptomatic until it has grown to a large size.

Pathology

Serous
  • Benign serous tumours are loculated, have a single layer of flattened or cuboidal epithelium and the absence of mitoses. Papillae are sometimes present on the external or internal surfaces. Examples of serous cystadenomas can be found at:
    http://www-medlib.med.utah.edu/WebPath/FEMHTML/FEM054.html - gross appearance
    http://www-medlib.med.utah.edu/WebPath/FEMHTML/FEM073.html - gross appearance
    http://www-medlib.med.utah.edu/WebPath/FEMHTML/FEM052.html - gross appearance
    http://www-medlib.med.utah.edu/WebPath/FEMHTML/FEM053.html -demonstrates internal papillae
    http://www-medlib.med.utah.edu/WebPath/FEMHTML/FEM051.html - illustrates external papillae
    http://www-medlib.med.utah.edu/WebPath/FEMHTML/FEM056.html - H and E staining
    http://pathweb.uchc.edu/eAtlas/GYN/1314.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1315.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1316.htm
    http://pathweb.uchc.edu/eAtlas/GYN/61.htm
    http://pathweb.uchc.edu/eAtlas/GYN/159.htm
    Histological analysis of borderline serous tumours reveals papillary cystic pattern, stratification, tufting, increased mitotic figures and cytologic atypia. H and E histology of borderline serous tumour can be viewed at:
    http://www-medlib.med.utah.edu/WebPath/FEMHTML/FEM055.html.

  • Malignant serous tumours are soft, multiloculated, partially cystic, partially solid tumours with friable papillae. Their capsule may be smooth or irregular or show papillary projections. Internal papillae are soft and tan in colour. Cyst fluid is clear, thin and colourless. The gross appearance of serous cystadenocarcinoma can be viewed at:
    http://pathweb.uchc.edu/eAtlas/GYN/463.htm
    Histological review of malignant serous tumours indicates significant stromal invasion. Calcifications (Psammoma bodies) are present in one-third of patients. Characteristic microscopic features include finger-like papillae with fibrovascular core, covered by multilayered cuboidal or columnar epithelium, hyperchromatic nuclei, prominent nucleoli, frequent mitoses, Psammoma bodies and desmoplasia (invasion of stroma with fibrosis). Examples can be viewed at:
    http://pathweb.uchc.edu/eAtlas/GYN/1927.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1928.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1929.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1097.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1098.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1099.htm
    http://www-medlib.med.utah.edu/WebPath/FEMHTML/FEM072.html -shows Psammoma bodies

    Mucinous

  • Benign mucinous tumours are larger than serous tumours, and may grow to an enormous size. They are usually unilocular cysts or may have a few septae, with a smooth external surface. The cyst fluid is slimy, yellow and clear. Mucinous tumours are the most heterogeneous group of epithelial tumours. Benign mucinous tumours have a single layer of tall, columnar cells and clear, mucin-producing cells, with a bland stroma. Microscopic images of mucinous cystadenoma can be viewed at:
    http://pathweb.uchc.edu/eAtlas/GYN/1930.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1931.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1932.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1933.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1310.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1311.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1312.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1313.htm

  • Borderline mucinous tumours have complex patterns, two to three cell layer stratification, cytological atypia and mitotic figures. Carcinoma is diagnosed when the stratification exceeds three cell layers or if there is a significant stromal invasion . Mucinous cystadenocarcinomas contain a smooth capsule, are cystic, multiloculated and large tumours (can be 50 cm in diameter). The cystic fluid is clear, yellow and sticky. The gross appearance of a mucinous cystadenocarinoma can be viewed at:
    http://pathweb.uchc.edu/eAtlas/GYN/530.htm
    Their microscopic appearance resembles intestinal adenocarcinoma. Multiple glands comprising mucin-containing cells are present, demonstrating nuclear atypia, hyperchromasia with prominent nuclei and desmoplasia.

    Borderline/LMP
    Borderline/LMP tumours are characterised by epithelial multilayering of more than 4 cell layers, and less than 4 mitoses per 10 high-power field, mild nuclear atypia, increased nuclear/cytoplasmic ratio, slight-to-complex branching of epithelial papillae and pseudopapillae, epithelial budding and cell detachment into the lumen and no destructive stromal invasion. Borderline mucinous tumours have similar gross morphology to their benign counterparts, cysts with smooth surfaces. The epithelial layer is characterised by stratification of 2-3 layers, nuclear atypia, enlarged nuclei and mitotic figures. Histological examples of borderline mucinous tumours can be found at:
    http://pathweb.uchc.edu/eAtlas/GYN/1934.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1935b.htm
    Approximately 25% of borderline tumours show cell proliferations on the outer surface only. Of these, 90% develop peritoneal implants, which can be invasive or non-invasive. Both have a similar appearance, glandular or papillary proliferations with cell detachments, sometimes Psammoma bodies, cellular atypia and desmoplastic fibrosis. However, epithelial cells infiltrate the stroma in the invasive implants.

    Brenner tumours
    Brenner tumours are solid or cystic, yellow-tan colour and firm upon gross examination. Histological examination of Brenner tumour reveals epithelial nests or cysts of cells, resembling urothelium, separated by a cellular, fibrous stroma (composed of spindle-like cells). The nuclei are relatively uniform, lacking pleomorphism, hyperchromasia or macronucleoli, and mitoses are not identified. There is a moderate amount of eosinophilic cytoplasm. Examples of Brenner tumours can be viewed at:
    http://pathweb.uchc.edu/eAtlas/GYN/1941b.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1942.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1943.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1299.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1300.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1301.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1302.htm

    Clear Cell Carcinoma
    Clear cell carcinoma accounts for 5-12% of ovarian adenocarcinomas. The gross appearance of clear cell carcinoma shows a smooth, lobulated external surface. These tumours are usually solid and firm, but can be cystic. They have a yellow-tan colour. Microscopic examination reveals cells arranged in tubules, nests or cysts, with clear, glycogen rich cytoplasm, sharply demarcated cell borders, and hyperchromatic, pleomorphic nuclei. Hobnail cells with nucleus standing on a stalk of cytoplasm are visible microscopically. Examples of clear cell carcinoma can be found using the following weblinks:
    http://pathweb.uchc.edu/eAtlas/GYN/1937.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1938.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1939.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1940.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1049.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1050.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1051.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1052.htm
    http://pathweb.uchc.edu/eAtlas/GYN/1053.htm

    Endometrioid Carcinoma
    Endometrioid carcinomas are solid, white, firm tumours with smooth or irregular surfaces. They may contain a cystic component and have areas of necrosis and haemorrhage. Histological analysis reveals glands, or glands mixed with solid areas, round-oval vesicular, clear nuclei with prominent nucleoli. Endometrioid carcinoma is indistinguishable from endometrial carcinoma. An example of endometrioid carcinoma can be found at:
    http://pathweb.uchc.edu/eAtlas/GYN/437.htm

    Mixed Mesodermal
    The gross appearance of mixed mesodermal ovarian tumours are exemplified in the following weblinks:
    http://pathweb.uchc.edu/eAtlas/GYN/134.htm
    http://pathweb.uchc.edu/eAtlas/GYN/135.htm
    http://pathweb.uchc.edu/eAtlas/GYN/136.htm
    They are usually large variegated lesions with necrotic and haemorrhagic regions, and may have adhesions. Microscopic examination reveals serous or endometrioid epithelial component displaying squamous differentiation. Stroma may comprise spindle cell or soft tissue differentiation including cartilage, skeletal muscle or smooth muscle.

  • Treatment

    The primary treatment of epithelial ovarian cancer is aggressive surgical tumour debulking, including total abdominal hysterectomy and bilateral salpingo-oophorectomy. Most women with ovarian epithelial tumours, except some stage Ia patients, receive chemotherapy. Postoperative treatment usually involves taxane-platinum combination chemotherapy; cisplatin or carboplatin with paclitaxel is the usual first-line treatment. High response rates, about 80%, are obtained, however most patients relapse, and other combination therapies fail. The mean disease-free interval for patients with stage III and IV disease is about 18 months. Only 20-30% of stage III and IV cases are long-term survivors. Postoperative intraperitoneal chemotherapy or external radiation therapy are used to treat patients with minimal residual disease. Clear cell adenocarcinoma is usually resistant to platinum-based chemotherapy. A strong association exists between ovarian mucinous tumours and appendiceal mucinous neoplasms. Consequently the appendix should be removed in patients with mucinous neoplasms. Repeat laparotomy or peritoneal lavage is required to remove gelatinous material in the persistent recurrences of Pseudomyxoma peritonei . Brenner tumours are cured with surgical resection. Prophylactic oophorectomy at an early age has significantly reduced the risk of coelomic epithelial cancer. Oral contraceptives have a protective effect against ovarian cancer in carriers of BRCA1 or BRCA2 mutations.

    Evolution

    Epithelial ovarian cancer initially spreads by direct seeding of the peritoneal surfaces, and is found on the underside of the diaphragm, paracolic gutters, bladder, cul-de-sac, surface of liver, mesentery and serosa of large and small bowel, omentum, uterus and paraaortic and pelvic lymph nodes. The tumour cells may remain confined to the surface of the coated abdominal viscera without penetrating it. They may also spread to the pleural cavity, lungs and groin lymph nodes. Mucinous tumours tend to form large masses, whereas serous tumours tend to distribute more diffusely, and are more often bilateral. Endometrioid and clear cell tumours usually invade locally and retroperitoneally. Sometimes mucus-secreting ovarian carcinomas fill the peritoneal cavity with a gelatinous neoplastic mass, referred to as pseudomyxoma peritonei .

    Prognosis

    The most important determinant for a favourable prognosis is diagnosis of ovarian carcinoma at an early stage. The prognosis of invasive epithelial ovarian cancer is poor, and relates to stage (see Tables 1 and 2), tumour grade and residual disease after surgery. The prognosis or early-stage ovarian invasive cancers and borderline tumours of all stages is significantly better. 5-year survival rates for patients with stage I disease are more than 90%, but less than 25% for advanced stage cancers. Patients with borderline tumours have an excellent prognosis. Age at diagnosis and the presence of invasive peritoneal implants are associated with a poorer prognosis in borderline tumours. The recurrence rate is 20%, with a mean time from diagnosis to relapse is 3.1 years in women with borderline serous tumours with non-invasive implants. However, borderline serous tumours with invasive implants have much higher recurrence rates of 32-45%, which occur much earlier (median time 24 months).

    Clear cell adenocarcinoma has a worse prognosis that the other histological subtypes as it is resistant to platinum-based chemotherapy. Some data suggests familial ovarian cancers have prolonged survival in comparison to the nonfamilial cases. In one study, patients with familial ovarian cancer exhibited a 67% 5-year survival, in comparison with a 17% 5-year survival in the nonfamilial ovarian cancer cases.

    STAGEDEFINITION
    Stage IGrowth limited to ovaries
    Stage IaGrowth limited to one ovary, no ascites, no tumour on external surface, capsule intact
    Stage IbGrowth limited to both ovaries, no ascites, no tumour on external surface, capsule intact
    Stage IcTumour either stage Ia or Ib, but with tumour on one or both ovaries, with capsule ruptured, with ascites present containing malignant cells, or with positive peritoneal washings
    Stage IIGrowth involving one or both ovaries with pelvic extension
    Stage IIaExtension and/or metastases to the uterus and/or tubes
    Stage IIbExtension to other pelvic tissues
    Stage IIcTumour either stage IIa or IIb, with tumour on the surface of one or both ovaries, but with capsule(s) ruptured, with ascites present containing malignant cells, or with positive peritoneal washings
    Stage IIITumour involving one or both ovaries with peritoneal implants outside the pelvis and/or positive retroperitoneal or inguinal nodes. Superficial liver metastases equal stage III. Tumour limited to the true pelvis but with histologically proven malignant extension to small bowel or omentum
    Stage IIIaTumour grossly limited to the true pelvis with negative nodes but with histologically confirmed microscopic seeding of abdominal peritoneal surfaces.
    Stage IIIbTumour involving one or both ovaries with histologically confirmed implants of abdominal peritoneal surfaces, none exceeding 2cm in diameter. Nodes are negative.
    Stage IIIcAbdominal implants >2cm in diameter and/or positive retroperitoneal or inguinal nodes.
    Stage IVGrowth involving one or both ovaries with distant metastases.

    Table 1 Definitions of the FIGO classification scheme for Staging Primary Ovarian Carcinoma (taken from Jones, 2000)

    STAGE NoPATIENTS TREATEDSURVIVAL3-yr (%)SURVIVAL 5-yr (%)
    I5,55987.582.1
    II3,36472.164.5
    III2,53047.038.1
    IV49220.714.0
    TOTAL11,94571.665.4

    Table 2 Survival Rates of Ovarian Carcinoma according to Disease Stage (adapted table from Jones, 2000)

    Cytogenetics

    Atlas Image
    Figure 1 Summary of karyotypic evolution in Ovarian carcinomas (taken from Hoglund et al., 2003)

    Cytogenetics morphological

    There is far more cytogenetic data available on ovarian carcinomas than for the other subtypes of ovarian tumours (germ cell tumours, sex-cord stromal tumours). At present, there are over 400 published karyotypes of ovarian carcinomas. The cytogenetic aberrations are non-random and complex. However, no pathognomonic rearrangements have been identified thus far. The karyotypes often show severe aneuploidy, with hypodiploid or near-triploid stemline chromosome numbers. The different subtypes of ovarian carcinoma show no marked cytogenetic differences, except seropapillary tumours more frequently display chromosome aberrations than the other subtypes. Complex chromosomal aberrations are present in invasive carcinomas, but not in benign or LMP tumours. The complexity of the karyotypes obtained from advanced tumours have obscured the initiating events in the pathogenesis of these tumours. Often normal karyotypes or simple cytogenetic aberrations were found in low-grade tumours. However, a correlation exists between karyotypic complexity and tumour grade. Simple chromosome changes (numerical changes only or a single structural rearrangement) were found in well-differentiated carcinomas, whereas complex karyotypes were found in poorly differentiated tumours. Patients with aberrant tumour karyotypes, particularly complex ones, were associated with short survival. Approximately 10-20% of ovarian carcinomas display homogeneously staining regions (hsr), although the loci they contain are unknown. However dmin are rarely observed. The most prevalent numerical changes are gains of chromosomes 1, 2, 3, 6, 7, 9, 12 and 20 losses of chromosomes 4, 8, 11, 13, 14, 15, 17 and 22. Structural rearrangements primarily involve deletions and unbalanced translocations involving 1p, 1q, 3p, 3q, 6q, 7p, 10q, 11p, 11q and 12q. In the review of 244 primary ovarian adenocarcinomas 201/244 tumours displayed clonal chromosomal abnormalities and hsr were identified in 20 cases. Using log-rank and proportional hazards regression analysis, it has been found that the presence of a chromosome breakpoint in any of 21 nonrandomly involved regions and breaks in 9 distinct regions (1p1, 1q2, 1p3, 3p1, 6p2, 11p1, 11q1, 12q2, and 13p1) were associated with reduced patient survival rate and time. Furthermore, only breakpoints within 1p1 and 3p1 retained independent, deleterious effects on survival and clinical variables associated with survival. In one review, 37% of serous LMP tumours displayed chromosomal anomalies, commonly trisomies of 7, 8 and 12. A much higher proportion, 91%, of invasive serous carcinomas of low-grade malignancy display clonal chromosomal abnormalities. A combination of karyotyping and microsatellite analyses identified a small deletion of 6q27, between D6S149 and D6S193, in both benign and advanced ovarian epithelial tumours, suggesting the presence of a putative tumour suppressor gene which is involved in the early events of the genesis of this tumour.

    Invasive serous and undifferentiated ovarian carcinomas have complex cytogenetic rearrangements, including amplification of oncogenes. Complex chromosomal anomalies are rarely found in mucinous and endometrioid carcinomas (mainly in advanced stages), and are never found in serous LMP tumours. Epithelial ovarian tumours are characterised by gains at 3q, 8q and 20q, often with high level amplification. Thus the cytogenetic profiles of ovarian carcinomas differ from that of ovarian granulosa cell tumours, trisomy 14 and monsomy 22 are rarely found in ovarian carcinomas. Chromosome 1 and 3 abnormalities are the commonest aberrations found in ovarian metastatic tumours. Cytogenetic investigation of 11 individuals with bilateral ovarian carcinoma showed identical baseline karyotypes, suggesting both tumours arise from the same transformed cell, rather than the tumours arising independently.

    46/52 ovarian carcinomas had complex karyotypes, often with a stemline chromosome number that was approaching near-triploid or hypodiploid. Chromosome losses of X, 22, 17, 13, 14 and 8, (lost in 15 imbalances

    Their analyses hypothesised that the temporal order of imbalances were as follows: 1q-, 6q-, +7 and +8q occurred early, -4, -8, +1q, +12 and +20 were intermediate imbalances, and the remaining imbalances were late events. It has been concluded that karyotypic evolution in ovarian carcinomas followed at least 2 cytogenetic pathways. The first pathway involved chromosomal gains of +7/+8q/+12 and was associated with low-stage and low-grade tumours. The second pathway involved chromosomal losses of 6q- and 1q- was found in tumours of moderate stage and grade. The early stages of karyotypic evolution result from the step-wise acquisition of changes resulting in Phase I tumours. Chromosome instability resulted in the transition to Phase II tumours, possibly as a result of extensive telomere crisis and breakage fusion breakage cycles, which is linked to imbalances characteristic of the 6q-/1q- pathway. Consequently, low-grade and borderline tumours cannot progress unless they have mixed-pathway features. The 6q-/1q- pathway was associated with triploidization. The 6q-/1q- pathway is instrumental in the progression of ovarian carcinomas. The proposed pathway of karyotypic evolution in ovarian carcinomas is summarised in Figure 1.

    A cohort of 114 ovarian neoplasms was analysed, including benign, borderline and invasive carcinomas by conventional and molecular cytogenetics. The chromosome abnormalities were categorised as follows:

  • Group 1: Abnormalities found in all subtypes. This included losses of chromosomes 6, 8, 10, 11, 15, 16, 17, 18, 19, 20, 21, 22 and X together with 6q24-qter deletions; and gains of chromosomes 1, 3, 5 and 12.
  • Group 2: Abnormalities present in malignant but not benign subtypes. This included losses of chromosomes 2, 7, 13 and 14, and gains of chromosome 4 and marker chromosomes.
  • Group 3: Abnormalities unique to invasive carcinomas such as loss of chromosome 4, 6q16-q24 deletions, gains of chromosomes 2, 7, 8, 9, 10, 16, 17, 18, 19, 20 and 21, and structural rearrangements of 3p, 3q, 13q and 21q.

    The presence of cytogenetic aberrations common to all subtypes suggests these tumours develop by progression.

    The main conclusions from cytogenetic investigations of ovarian epithelial tumours are as follows:

  • Nonrandom breakpoints in ovarian adenocarcinoma do not occur independently.
  • Breakpoints in 1p3 and 11p1 are early events, and associated with poor prognosis
  • Breakpoints in 1p1, 3p1 and 1q2 distinguish a class of ovarian tumours, and breakpoints at 1p1 and 3p1 are associated with a poor prognosis.
  • Cytogenetics molecular

    Interphase cytogenetics demonstrated a high frequency of gain of copy number of 20q13.2 (70%) and cyclin D1 (CCND1 at 11q13, 72%) which were associated with poor prognosis. Another study addressing amplification of 20q12-q13.2 using a series of FISH probes in 24 sporadic and 7 hereditary ovarian carcinomas found amplification of at least one of the regions in 54% of sporadic cases and all of the hereditary cases, and amplification of AIB1 (20q12), a steroid receptor coactivator correlated with poor survival.Online access to summaries of the recurrent DNA copy number amplifications and losses identified by CGH in ovarian epithelial neoplasms (and other tumour entities) can be viewed at http://www.helsinki.fi/cmg/cgh_data.html, and undergo regular updates. The criteria for recurrent losses and gains employed were as follows. For losses, 10% of the cases should have the loss, and there must be at least 3 aberrant cases. Highly frequent aberrations which do not meet the criteria of 10% of cases or 3 cases are indicated by parentheses-such as 1p21-p31. Recurrent amplicons were defined as at least 3 cases and >5% frequency display the amplicon. The recurrent losses and gains are summarised in Table 4.

    Ovarian Neoplasm SubtypeLossAmplificationPercentage (number of cases)
    Ovarian Cancer(1p21-p31)7 (5/72)
    1p34.1-p34.311 (5/44)
    1q7 (5/71)
    2p15p224 (1/24)
    2q22-q245 (1/20)
    3cen-q234 (1/24)
    3q25-q2713 (6/47)
    4q21-q3216 (30/184)
    4q32-qter16 (15/91)
    4q32-qter16 (15/91)
    5q12-q2316 (30/184)
    6p21 7 (3/144)
    6q13-qter5 (1/20)
    6q16-qter13 (23/184)
    7q367 (2/27)
    8p21-pter17 (32/184)
    8q4 (1/24)
    8q22-qter18 (8/44)
    9p10 (16/159)
    9p244 (1/27)
    9q13 (17/136)
    10p154 (1/27)
    10q11-qter17 (10/59)
    12p1219 (9/47)
    12p129 (4/47)
    12q24-qter10 (14/140)
    13q12-q2118 (24/135)
    13q21-q3212 (18/160)
    16p13 (12/93)
    16q23 (24/184)
    17p20 (37/184)
    17q11.2-q3223 (31/137)
    17q21-qter6 (3/47)
    18p11.34 (1/27)
    18q12-qter18 (33/184)
    19p23 (10/44)
    19q16 (11/69)
    21q10 (12/116)
    22q18 (17/93)
    Xp19 (35/184)
    Xq19 (9/47)
    Xq11.2-q2113 (12/89)
    Xq21-qter24 (16/68)
    Primary epithelial ovarian cancer4p15.218 (5/28)
    4q23-q2418 (5/28)
    4q26-q2718 (5/28)
    5q1414 (4/28)
    5q1514 (4/28)
    9p22-p2411 (3/28)
    9q22-q3118 (5/28)
    13q1414 (4/28)
    13q31-q3221 (6/28)
    14q24.3-q3114 (4/28)
    15q21.125 (7/28)
    18q2111 (3/28)
    Ovarian Inherited (BRCA1 & BRCA2)1q32-qter10 (2/20)
    3q26.1 10 (2/20)
    5p5 (1/20)
    6p22-p2410 (2/20)
    6q21-q225 (1/20)
    6q25-qter15 (3/20)
    8p2340 (8/20)
    8q23-q24.130 (6/20)
    12p5 (1/20)
    12q13-q215 (1/20)
    18p11.2-pter15 (3/20)
    18q21-qter20 (4/20)
    20p5 (1/20)
    Xp15 (3/20)
    Xq12-2115 (3/20)
    Ovarian cancer, sporadic & inherited1p34-p3625 (4/18)
    4q31.3-q3519 (3/16)
    9q31-q3440 (8/16)
    10q23-q2625 (4/16)
    11q23-q2519 (3/16)
    16p19 (3/16)
    16q22-q2438 (6/16)
    17p19 (3/16)
    1919 (3/16)
    22q12-q1325 (4/16)
    Xp19 (3/18)

    Table 4 Recurrent amplifications and losses in epithelial ovarian tumours, including hereditary neoplasms (data taken from http://www.helsinki.fi/cmg/cgh_data.html)

    Other studies have identified gain of chromosome 8 in 1/10 ovarian carcinomas.

    A study of 31 primary ovarian carcinomas in Chinese women by CGH identified several non-random changes in copy number including gains of 3q (17 cases, 55%) with a minimum region of gain of 3q25-q26, 8q (16 cases, 52%), 19q (12 cases, 39%), Xq (11 cases, 35%), 1q (10 cases, 32%), 12p12-q13 (10 cases, 32%), 17q (10 cases, 32%) with a minimum region of gain at 17q21, and 20q (9 cases, 29%); together with losses of 16q (9 cases, 29%), 1p (7 cases, 23%), 18q (7 cases, 23%) and 22 (7 cases, 23%). High copy number amplifications were observed at 3q25-q26 (4 cases), 8q24 (3 cases) and 12p11.2-q12 (3 cases). The commonest imbalances detected by CGH of epithelial neoplasms were gain of 3q25-26, gain of 8q24, loss of 16q, and loss of 17pter-q21. 12p gains were seen in 8/44 cases, which has been reported previously in both ovarian and testicular germ cell tumours. Another study by Hauptmann et al., 2002 using CGH to analyse ovarian carcinomas identified frequent gains of 3q, 6p, 7, 8q and 20, together with losses of 4q, 6q, 12q, 13q and 16q, which have supported the available cytogenetic data.

    CGH was used to screen a mucinous ovarian carcinoma and a Brenner tumour coexisting in different ovaries of the same female. Amplification of 12q14-q21 was identified in both tumours, in the presence of other copy number changes, 4 such changes in the Brenner tumour and 6 in the mucinous carcinoma.

    Correlation of CGH data with Clinical data

    In a large study of 106 primary ovarian carcinomas, the CGH findings were correlated with clinical parameters such as tumour grade of differentiation. 103 tumours displayed imbalances. Amplifications of 8q, 1q, 20q, 3q and 19p were frequent findings present in 69-53% of the tumours. Underrepresentations of 13q, 4q and 18q were also common, present in 54-50% of cases. Underrepresentation of 11p and 13q and overrepresentation of 8q and 7q correlated with undifferentiated ovarian carcinoma, whereas 12p underrepresentation and 18p overrepresentation were more commonly associated with well-differentiated and moderately differentiated tumours. These findings corroborate other CGH studies including.

    A CGH study of a cohort of 12 ovarian clear cell carcinomas revealed similarities to the data of other subtypes of epithelial neoplasms, such as gains of 8q and 17q and losses of 19p. They also correlated their findings with disease status (i.e. disease free, recurrent disease, or death from disease). DNA copy number changes present in over 20% of cases included overrepresentation of 8q11-q13, 8q21-q22, 8q23, 8q24-qter, 17q25-qter, 20q13-qter and 21q22; and underrepresentation of 19p. Overrepresentation of 8q11-q13, 8q21-q22, 8q23, 8q24-qter was more common in disease-free patients than in those with recurrent disease or who had died. Conversely, overrepresentation of 17q25-qter, 20q13-qter was more frequent in patients with recurrent disease or non-survivors, than in disease-free patients. This data suggests ovarian clear cell carcinoma develop along 2 cytogenetic pathways.

    In a study correlating CGH genomic imbalances with clinical endpoints in 60 ovarian carcinomas, the following associations were found:

  • Loss of chromosome 4 with high-grade tumours
  • Gains of 3q26-qter, 8q24-qter and 20q13-qter and low-grade and low-stage tumours
  • Deletion of 16q24 and >7 independent genomic imbalances and reduced survival times
  • Tumour grade correlated better with genomic progression than clinical stage.

    CGH findings of Sporadic and Hereditary Ovarian Carcinoma

    CGH profiles were compared from sporadic and hereditary (3 BRCA1 and 1 BRCA2 mutation carriers) ovarian cancers. The commonest imbalance included amplification of 8q22.1-qter (66.6%), 1q22-32.1 (41.1%), 3q (75%) and 10p (33.2 %), and deletion of 9q (41.6%) and 16q21-q24 (33.3%). Deletions of 9q were found in all 3 BRCA1 carriers and 2/8 sporadic tumours, and deletions of 19 were found in 2/3 BRCA1 carriers and none of the sporadic cases. These findings suggest preferential somatic losses of chromosome 9 and 19 in BRCA1 mutation carriers. In contrast, another study identified extensive similarity by CGH between sporadic and hereditary ovarian carcinomas, except for 2q24-q32. CGH analysis of a further 36 hereditary tumours found the majority of imbalances to be similar to that of sporadic tumours (Gains: 8q23-qter, 3q26.3-qter, 11q22, 2q31-32; losses: 8p21-pter, 16q22-qter, 22q13, 12q24, 15q11-15, 17p12-13, Xp21-22, 20q13, 15q24-25, 18q21). However some imbalances were identified that were specific to hereditary tumours, including deletions of 15q11-15, 15q24-25, 8p21-pter, 22q13 and 12q24, and gains of 11q22, 13q22 and 17q23-35. Deletions of 15q11-15 and 15q24-25 were found in 16/36 and 12/36 cases respectively which implicated hRAD51 and other tumour suppressor genes in these loci in the genesis of hereditary ovarian cancer.

  • Genetics

    Note

    INHERITED PREDISPOSITION

    As mentioned in the Aetiology section, genetic factors are the most important risk factor for ovarian epithelial carcinoma. Having 1 or 2 first-degree relatives with ovarian cancer increases the lifetime risk to 3-5% and 39% respectively. Three hereditary syndromes in which familial aggregation of ovarian carcinoma occurs have been described:

  • Hereditary breast-ovarian cancer syndrome: clusters of breast and ovarian cancer among first- and second-degree relatives
  • Hereditary nonpolyposis colorectal cancer syndrome, HNPCC, or Lynch Cancer Family syndrome II): ovarian cancer develops in a proband whose close relatives have had cancers of the colon, breast, ovary, endometrium, urinary tract, uterine and other malignancies.
  • Site-specific ovarian cancer syndrome of unknown origin in which two or more first-degree relatives have ovarian cancer

    All 3 patterns of familial ovarian cancer are consistent with autosomal-dominant transmission of one or more genes responsible for the development of >1 cancers, with incomplete penetrance and variable expression. The age of diagnosis of hereditary epithelial ovarian cancer is approximately 10 years earlier than its sporadic counterpart.

    Breast-Ovarian Syndrome
    Of the about 10% of ovarian epithelial cancers thought to have a hereditary component, 90% are associated with breast-ovarian syndrome. This syndrome is associated with two genes, BRCA1 at 17q21, and BRCA2 at 13q12.3 (see below), which are involved in DNA repair and transcription regulation. Mutations are distributed throughout the entire coding regions of BRCA1 and BRCA2, and most result in truncation of the protein. Germline mutations in BRCA1 account for about 80% of hereditary breast-ovarian cancers. Germline mutations of BRCA2 account for about 10-35% of familial ovarian cancers. BRCA1 is associated with a 26% cumulative risk for ovarian cancer for most mutation carriers, and a much higher risk, 85%, in a small subset. Women with a germline BRCA1 mutation have an about 40% risk of developing ovarian cancer by 70 years of age. BRCA2 increases susceptibility to a smaller degree. The lifetime risk for developing ovarian cancer in BRCA2 mutation carriers is 27%. However the risks of developing ovarian cancer associated with germline mutations of BRCA1 and BRCA2 vary according to the population studied. A study by revealed a lifetime risk of ovarian cancer of 40-60% for BRCA1 mutation carriers, whereas another one found a 25-30% risk for BRCA1 mutation carriers. Approximately 1/4000 in the general population has a mutation of BRCA1, although some populations have much higher incidences, for example the Ashkenazi Jews. Patients with breast cancer who had BRCA1 or BRCA2 mutations had a tenfold increased risk of developing ovarian cancer.

    The variable penetrance of BRCA1 suggests that other genetic and non-genetic factors contribute to the pathogenesis in these individuals. One such modifier is a VNTR polymorphism, 1-kb downstream of HRAS. BRCA1 carriers with rare alleles of the VNTR had an 2.11 increased risk of developing ovarian cancer compared with the common alleles (p=0.015).

    about 50% of familial ovarian cancers are not associated with germline BRCA1 or BRCA2 mutations. Linkage and LOH analysis has suggested a susceptibility gene for familial ovarian cancer at 3p22-p25. LOH of 3p33-p25 is higher (52%) in non-BRCA1/BRCA2 familial ovarian cancers than in the BRCA1 (29.7%) group.

    HNPCC
    Mutations of the mismatch repair genes (MMR) including MLH1, MSH2 and MSH6 are present in HNPCC syndrome (Lynch 2 Syndrome). This represents the second most common type of ovarian cancer with a hereditary component.

    Site-Specific Ovarian Cancer Syndrome
    The least common of the familial ovarian cancers is the site-specific ovarian cancer syndrome, in which ovarian cancer is the dominant cancer. It has been suggested that site-specific ovarian cancer is a variant of breast-ovarian syndrome attributable to mutation in either BRCA1 or BRCA2, and not a distinct clinical entity.

    Early onset ovarian carcinoma (

  • Genes Involved and Proteins

    Note

    In addition to involvement of germline mutations of BRCA1, BRCA2 and the mismatch repair genes in the predisposition of ovarian epithelial tumours (see Genetics: Inherited Predisposition section), many studies have investigated somatic changes at specific loci. The somatic aberrations are summarised below according to whether the studies involved allelotyping or analysing specific genes (which is further subdivided into oncogenes and tumour suppressor genes).

  • PI3K/AKT2
    Cytogenetic and loss of heterozygosity (LOH) studies have implicated many regions of the genome in the pathogenesis of ovarian cancer. Numerous allelotype studies have been performed on ovarian carcinomas and identified frequent losses of 1p, 4p, 5q, 6p, 6q, 7p, 8p, 8q, 9p, 9q, 11p, 11q, 12p, 12q, 13q, 14q, 15q, 16p, 16q, 17p, 17q, 18q, 19p, 21q, 22q and Xp. However most of these studies examined ovarian serous adenocarcinoma. From the few studies that have analysed LOH of benign and LMP tumours, LOH is rarely found at most loci, with the exception of the X chromosome in LMP tumours. LOH analysis of early-stage malignant and borderline ovarian tumours displayed similar LOH patterns suggesting that malignant ovarian tumours may develop from benign and borderline tumours. Frequent allelic losses are found at 5p15.2, 5q13-21, 6p24-25, 6q21-23, 6q25.1-27, 7q31.1, 11p15.5, 11p13, 11q22, 11q23.3-qter, 17p13.3 and 17p11.2, suggesting the presence of tumour suppressor genes involved in ovarian carcinoma. Microcell-mediated chromosome transfer of normal chromosome 11 and 17 confirmed the presence of tumour suppressor gene(s) on these chromosomes. Complete suppression of tumourigenicity was obtained by transfer of chromosome 11, whereas reduced in vivo and in vitro growth rates together with increased latency period were obtained by the transfer of chromosome 17. Furthermore transfer of 17p11.2 had the same effect as transfer of the entire chromosome. Microsatellite analysis has suggested the presence of a tumour suppressor gene at 22q11-q12 (between D22S301 and D22S304). This was also supported by microcell-mediated chromosome transfer of chromosome 22 into ovarian carcinoma cell line SKOV3 which resulted in complete abrogation of anchorage-independent growth and a dramatic reduction of in vitro doubling times and tumourigenicity in nude mice.

    The pattern of allelic loss differs according to the histological subtypes of epithelial ovarian cancer. Clear cell adenocarcinoma predominantly demonstrates LOH of 1p, 19p and 11q. Serous adenocarcinoma demonstrates allelic losses in >50% of cases of 1p, 4p, 5q, 6p, 8p, 9q, 12q, 13q, 15q, 16p, 17p, 17q, 18p, 18q, 19p, 20p and Xp. Endometrioid adenocarcinoma frequently demonstrated LOH of 7p, and mucinous adenocarcinoma demonstrated recurrent LOH at 17p13.1. LOH analysis using RFLP markers in 6q24-q27 demonstrated allelic loss at a few or all loci in 17/33 ovarian serous tumours, 1/15 ovarian mucinous tumours, and 2/12 ovarian clear cell tumours. Allelic loss of 1p31 has been found in about 40% of ovarian carcinomas, where the maternally imprinted tumour suppressor gene ARH1(NOEY2) resides. Approximately 1/3 of epithelial ovarian tumours of all stages demonstrate LOH of 9p. 69% of 78 ovarian epithelial tumours displayed LOH of 17p13.1 where TP53 is located. Allelic loss at 10q23.3 flanking PTEN and within PTGN have been found in 45% of ovarian epithelial cancers (n=68). Loss of PTEN expression was associated with elevated phosphorylated AKT levels. No microsatellite instability (MSI) was apparent among the 23 benign cystadenomas and 31 LMP ovarian tumours examined using 69 microsatellite markers. Thus these findings suggest MSI is not a pathogenic mechanism in the development of LMP tumours, and abnormalities of the DNA mismatch repair mechanisms are not involved. In contrast, about one-third of endometrioid carcinomas and up to 40% of serous LMP tumours display MSI, although in serous LMP tumours the MSI is low level.

    LOH of 3p14.2, 11p15.5, 11q23.3, 11q24, 16q24.3 and 17p13.1 are more frequent in advanced than lower stage tumours. LOH of 3p14.2 correlated with tumour metastasis, whereas LOH at 11p15.5 and 11q23.3 were associated with reduced survival. LOH of 11q22.3 was associated with reduced survival and a serous histology, meanwhile LOH of 11q24-25 correlated with a higher tumour stage, serous histology, presence of residual tumour, but not with survival. LOH of 1p36 is associated with poor histological grade.

  • Growth Factor Receptors
    Alterations in tumour suppressor genes such as P53, RB1, ARH1 (NOEY2), BRCA1 are involved in ovarian carcinogenesis.

  • Oncogenes involved in endometrioid carcinoma
    Allelic deletions of 17p or P53 mutations occur frequently in ovarian carcinoma. P53 mutations are found in about 50-80% of tumours when analysed by complete gene sequencing. LOH of P53 is also a frequent finding in ovarian carcinomas, ranging from 30% to 80%. P53 mutations have been found in ovarian carcinoma and borderline ovarian tumours. Invasive serous and undifferentiated ovarian carcinomas are characterised by P53 mutations with protein accumulation, extensive allelic loss of chromosome 17 and complex cytogenetic aberrations. Functional wild-type P53 is required for chemo- and radio-sensitivity due to its role in apoptosis. Thus mutation of P53 followed by loss of the wild-type results in resistance to therapy. Of the ovarian neoplasms that express nuclear P53, 90% of them have mutations of P53 which increases the half-life of the P53 protein. 50% of advanced ovarian carcinomas have overexpressed or mutant P53 which correlates with high grade and poor survival, but not with chemoresponsiveness. However, P53 does not appear to be involved in the pathogenesis of clear cell adenocarcinoma.

    Homozygous deletions or intragenic mutations of CDKN2A (p16INK4A) are also found in ovarian epithelial tumours. CDKN2A encodes an inhibitory protein of cyclin-dependent kinase 4. The CDKN2A complex blocks phosphorylation of the Retinoblastoma (RB) protein. Phosphorylation of the RB protein is a prerequisite for cells to enter the S phase of the cell cycle. Thus CDKN2A is a negative regulator of the cell cycle.

  • CSF1R (formerly fms, macrophage colony stimulating factor receptor), is expressed in many ovarian cancers, but not benign ovarian tumours or normal ovarian surface epithelium. Amplification of CSF1R has been reported in 3-5% of ovarian cancers.No expression of GATA4, a transcription factor gene located at 8p23.1, was found in the majority of serous carcinomas, whereas it is expressed in most mucinous carcinomas, suggesting that these tumour types develop along discrete pathogenic pathways.

    Reduced expression of RNASET2 (RNASE6PL), located at 6q27, was found in 30% of ovarian cancers. Transfection of RNASET2 cDNA into ovarian cancer cell lines suppressed tumourigenicity, suggesting it to be a candidate tumour suppressor gene.

    Somatic mutations of BRCA1 and BRCA2 have not been found in sporadic ovarian neoplasms, however allelic losses including 17q21, were BRCA1 is located, were common. This suggests that additional tumour suppressor genes are required in the molecular aetiology of sporadic tumours, one proximal to BRCA1, the other on 17p.

    Alterations in oncogenes KRAS, MYC and ERBB2 are frequently involved in ovarian carcinogenesis.

    KRAS mutations are found in 30% of ovarian carcinomas, and are frequently observed in mucinous adenoma and thus may be an early event in the pathogenesis of ovarian mucinous tumours. KRAS mutations are present in 40-50% of mucinous LMP tumours and mucinous carcinomas, and also in one-third of serous LMP tumours. Amplification of KRAS has been reported in 3-5% of ovarian cancers. In one study, KRAS2 amplification occurred in 2/53 of ovarian epithelial tumours, (6 borderline serous, 2 low grade serous, 31 high grade serous; 4 low grade mucinous; 2 low grade endometrioid, 8 high grade endometrioid), and only in the aggressive types (2 high grade serous tumours). A mutation in codon 12 of KRAS has been identified in small cell carcinoma.

    HRAS acquires transforming activity either as a result of substitution mutations or by increased expression of the normal gene. Mutated HRAS lack GTPase activity, resulting in dysregulation of cell growth.

    Abnormal cell signalling mediated by protein kinases can result from alterations of the growth factor receptors in ovarian epithelial neoplasms. These include:

  • ERBB2 (HER2/Neu) receptor which is amplified and overexpressed in 9-30% of ovarian cancers. The ERBB2 oncogene located at 17q21 encodes a membrane receptor that binds a glycoprotein similar to transforming growth factor-a and is correlated with poor survival of patients.
  • ECGF1, the platelet-derived endothelial growth factor, shows significantly higher levels in primary epithelial ovarian tumours and was more abundant at the higher stages (III and IV than lower stages), also more prevalent in the mucinous than in the serous adenocarcinomas.
  • EGFR which encodes the transmembrane receptor for epidermal growth factor is expressed by most advanced carcinomas and is associated with poor prognosis.

    Amplification of MYC oncogene, 8q24, occurs in 10-20 % of ovarian cancers, and in about one-third of advanced ovarian carcinomas. MYC amplification is more frequently found in the serous subtypes than the mucinous subtypes. MYC encodes a DNA-binding nuclear-associated protein that regulates cell proliferation. Dividing cells have increased amounts of nuclear c-myc, whereas quiescent cells express negligible quantities. MYC amplification is often indicative of biologically aggressive tumours. MYC amplification was not associated with prognosis or survival. Significantly higher levels of p62c-myc were found in serous papillary ovarian carcinoma. LMP tumours expressed MYC at values intermediate between that of normal ovary tissue and carcinoma.

    Amplification, altered expression, and malfunction of several protein kinases and phosphatases are involved in the pathogenesis of ovarian epithelial neoplasms, in particular the phosphatidylinositol 3-kinase (PI3K) pathway. Increased PI3K activity is important in the growth and dissemination of ovarian cancer cells. The PIK3CA gene which encodes the catalytic subunit of PI3K, and its downstream effector AKT2 are amplified in primary ovarian tumours. Overexpression of AKT2 is found in high-grade and late-stage tumours. Mutation and/or down-regulation of the PI3K phosphatase PTEN/MMAC1 are frequently observed in ovarian endometrioid carcinomas. AKT2 mediates some of the transforming signals of RAS and SRC which are mutated and overexpressed/activated respectively in late-stage tumours. Downregulation of the cGMP-dependent protein kinase PKG and upregulation of MAP2K6 (MEK6) were significantly correlated with the genesis of ovarian cancer. Amplification of AKT2 has been reported in 3-5% of ovarian cancers.

    Expression Profiling
    Amplification of other oncogenes such as FGF3 (formerly INT2) and MDM2 have been reported in 3-5% of ovarian cancers. As mentioned in the Molecular Cytogenetics section, high level amplification of 20q12-q13.2 is a frequent finding in ovarian carcinomas, and a gene located at 20q11.2-12, TGIF2, was amplified and over-expressed in 14 ovarian cancer cell lines. EIF5A2 is a candidate oncogene for the 3q25-q26 amplification in ovarian carcinomas. Overexpression of the Kallikrein gene, KLK4, located at 19q13.4, has been found in 69/147 ovarian tumours and is indicative of a poor prognosis. NME1 is thought to have a role in ovarian neoplastic process. Elevated levels of inhibin are found in most postmenopausal women with mucinous ovarian cancers.

    Overexpression of BCL2 is present in about 90% of endometrioid carcinomas, and MSI is present in about one-third of cases, as has been described in endometrioid endometrial carcinomas. Overexpression of P53, EGFR, ERBB2 and ERBB3 was also detected in ovarian endometrioid carcinoma.

    Expression microarrays were used to compare differential expression between 7 early stage ovarian carcinomas and 7 late stage ovarian carcinomas, and showed that several genes are aberrantly regulated to the same extent in both groups. Genes which function in cell-cell interaction such as cadherin 11 (CDH11), cadherin 2 (CDH2) and nidogen (NID) were downregulated in most tumours. Genes involved in invasion and metastasis such as matrilysin (MMP7), gelatinase (MMP9), matrix metalloproteinase 10 and 12 were upregulated in most tumours.

    Several other expression profiling studies have been undertaken which identified differentially expressed genes between serous and mucinous carcinomas; and also identified differences in gene expression during progression of ovarian carcinoma.

  • Bibliography

    Pubmed IDLast YearTitleAuthors
    105492711999Amplification of C-MYC as the origin of the homogeneous staining region in ovarian carcinoma detected by micro-FISH.Abeysinghe HR et al
    113140332001Cloning and characterization of a senescence inducing and class II tumor suppressor gene in ovarian carcinoma at chromosome region 6q27.Acquati F et al
    105646761999Mutational spectra of PTEN/MMAC1 gene: a tumor suppressor with lipid phosphatase activity.Ali IU et al
    114269682001Allele loss on chromosome 1p36 in epithelial ovarian cancers.Alvarez AA et al
    105185421999BRCA1-associated growth arrest is RB-dependent.Aprelikova ON et al
    91621971996Overrepresentation of 3q and 8q material and loss of 18q material are recurrent findings in advanced human ovarian cancer.Arnold N et al
    22275451990c-myc amplification in ovarian cancer.Baker VV et al
    76573931995Molecular alterations of the AKT2 oncogene in ovarian and breast carcinomas.Bellacosa A et al
    22985051990Chromosome aberrations in metastatic ovarian cancer: relationship with abnormalities in primary tumors.Bello MJ et al
    82968291994The p53 tumor suppressor gene frequently is altered in gynecologic cancers.Berchuck A et al
    93469401997Interaction between the adhesion receptor, CD44, and the oncogene product, p185HER2, promotes human ovarian tumor cell activation.Bourguignon LY et al
    84069991993Increased accumulation of p53 protein in cisplatin-resistant ovarian cell lines.Brown R et al
    79039481993Familial ovarian cancer.Buller RE et al
    97864541998Inhibin and ovarian cancer.Burger HG et al
    107333451999Characteristics of EGFR family-mediated HRG signals in human ovarian cancer.Campiglio M et al
    115663432001Suppression of tumorigenicity in human ovarian carcinoma cell line SKOV-3 by microcell-mediated transfer of chromosome 11.Cao Q et al
    89345441996DNA sequence analysis of exons 2 through 11 and immunohistochemical staining are required to detect all known p53 alterations in human malignancies.Casey G et al
    102084171999Methylation of the BRCA1 promoter region in sporadic breast and ovarian cancer: correlation with disease characteristics.Catteau A et al
    91159671997Analysis of loss of heterozygosity and KRAS2 mutations in ovarian neoplasms: clinicopathological correlations.Chenevix-Trench G et al
    14096331992AKT2, a putative oncogene encoding a member of a subfamily of protein-serine/threonine kinases, is amplified in human ovarian carcinomas.Cheng JQ et al
    86063791996Potential role of the inactivated X chromosome in ovarian epithelial tumor development.Cheng PC et al
    84857261993Human epithelial ovarian cancer allelotype.Cliby W et al
    92167251997Karyotypic analysis of 32 malignant epithelial ovarian tumors.Deger RB et al
    118943922001[Phenotype--genotype--correlation in ovarian neoplasia].Diebold J et al
    10727982200020q13 and cyclin D1 in ovarian carcinomas. Analysis by fluorescence in situ hybridization.Diebold J et al
    83063181994Evidence of functional RB protein in epithelial ovarian carcinomas despite loss of heterozygosity at the RB locus.Dodson MK et al
    92914301997Reduced expression of retinoblastoma gene product (pRB) and high expression of p53 are associated with poor prognosis in ovarian cancer.Dong Y et al
    84606341993Genetic linkage analysis in familial breast and ovarian cancer: results from 214 families. The Breast Cancer Linkage Consortium.Easton DF et al
    78255871995Breast and ovarian cancer incidence in BRCA1-mutation carriers. Breast Cancer Linkage Consortium.Easton DF et al
    94645371998A one centimorgan deletion unit on chromosome Xq12 is commonly lost in borderline and invasive epithelial ovarian tumors.Edelson MI et al
    94972461998Genetic heterogeneity and penetrance analysis of the BRCA1 and BRCA2 genes in breast cancer families. The Breast Cancer Linkage Consortium.Ford D et al
    18745721991Pooled analysis of 3 European case-control studies of epithelial ovarian cancer: III. Oral contraceptive use.Franceschi S et al
    96672591998Sequence analysis of BRCA1 and BRCA2: correlation of mutations with family history and ovarian cancer risk.Frank TS et al
    26472921989Association of Ki-ras with amplified DNA sequences, detected in human ovarian carcinomas by a modified in-gel renaturation assay.Fukumoto M et al
    113214882001Detection of numerical aberration in chromosome 17 and c-erbB2 gene amplification in epithelial ovarian cancer using recently established dual color FISH.Fukushi Y et al
    103290461999Inhibin subunit gene expression in ovarian cancer.Fuller PJ et al
    22275641990Chromosome abnormalities in human epithelial ovarian malignancies.Gallion HH et al
    106323391999Prognostic significance of p53 expression in advanced-stage ovarian serous borderline tumors.Gershenson DM et al
    113258562001Isolation of a novel candidate oncogene within a frequently amplified region at 3q26 in ovarian cancer.Guan XY et al
    128334572003Distribution of HER-2 overexpression in ovarian carcinoma tissue and its prognostic value in patients with ovarian carcinoma: from the Danish MALOVA Ovarian Cancer Study.Høgdall EV et al
    128106742003Ovarian carcinoma develops through multiple modes of chromosomal evolution.Höglund M et al
    22664601990c-erbB-2 oncogene expression in ovarian cancer.Haldane JS et al
    121521632002Genetic alterations in epithelial ovarian tumors analyzed by comparative genomic hybridization.Hauptmann S et al
    82064021994p53 expression in epithelial ovarian neoplasms: relationship to clinical and pathological parameters, Ki-67 expression and flow cytometry.Henriksen R et al
    107417112000In vivo and in vitro ovarian carcinoma growth inhibition by a phosphatidylinositol 3-kinase inhibitor (LY294002).Hu L et al
    82614571994Mutation of K-ras protooncogene is associated with histological subtypes in human mucinous ovarian tumors.Ichikawa Y et al
    85433151996Ovarian small cell carcinoma with K-ras mutation: a case report with genetic analysis.Idei Y et al
    110061162000Amplification and overexpression of TGIF2, a novel homeobox gene of the TALE superclass, in ovarian cancer cell lines.Imoto I et al
    111180612000Differential gene expression between normal and tumor-derived ovarian epithelial cells.Ismail RS et al
    85214101995Genetic analysis of benign, low-grade, and high-grade ovarian tumors.Iwabuchi H et al
    111040252000Comparison of comparative genomic hybridization and interphase fluorescence in situ hybridization in ovarian carcinomas: possibilities and limitations of both techniques.Jacobsen A et al
    82754571993Cytogenetic studies of epithelial ovarian carcinoma.Jenkins RB et al
    82052891994Epidemiology and primary prevention of cancers of the breast, endometrium, and ovary. A brief overview.Kelsey JL et al
    111699352001Comparative genomic hybridization detects genetic imbalances in primary ovarian carcinomas as correlated with grade of differentiation.Kiechle M et al
    79592841994Recurrent cytogenetic aberrations and loss of constitutional heterozygosity in ovarian carcinomas.Kiechle-Schwarz M et al
    14292091992High incidence of p53 gene mutation in human ovarian cancer and its association with nuclear accumulation of p53 protein and tumor DNA aneuploidy.Kihana T et al
    83063171994Loss of heterozygosity on chromosome 13 is common only in the biologically more aggressive subtypes of ovarian epithelial tumors and is associated with normal retinoblastoma gene expression.Kim TM et al
    83609341993Spectrum of mutation and frequency of allelic deletion of the p53 gene in ovarian cancer.Kohler MF et al
    4379121979Homogeneously staining regions on marker chromosomes in malignancy.Kovacs G et al
    111753422000Functional evidence for an ovarian cancer tumor suppressor gene on chromosome 22 by microcell-mediated chromosome transfer.Kruzelock RP et al
    104996291999Gains of 1q21-q22 and 13q12-q14 are potential indicators for resistance to cisplatin-based chemotherapy in ovarian cancer patients.Kudoh K et al
    85063421993p53 gene mutations and protein accumulation in human ovarian cancer.Kupryjańczyk J et al
    113953872001Frequent loss of PTEN expression is linked to elevated phosphorylated Akt levels, but not associated with p27 and cyclin D1 expression, in primary epithelial ovarian carcinomas.Kurose K et al
    76051681995Familial occurrence of small-cell carcinoma of the ovary.Lamovec J et al
    16145221992Cancer. p53, guardian of the genome.Lane DP et al
    111040332000Loss of heterozygosity at chromosomes 3, 6, 8, 11, 16, and 17 in ovarian cancer: correlation to clinicopathological variables.Launonen V et al
    85362361995p53 mutation in epithelial ovarian carcinoma and borderline ovarian tumor.Lee JH et al
    113245011997Overexpression of p53, EGFR, c-erbB2 and c-erbB3 in endometrioid carcinoma of the ovary.Leng J et al
    9032491977Chromosomes and cancer.Levan A et al
    94508581998Is hereditary site-specific ovarian cancer a distinct genetic condition?Liede A et al
    96799571998AKT2, a member of the protein kinase B family, is activated by growth factors, v-Ha-ras, and v-src through phosphatidylinositol 3-kinase in human ovarian epithelial cancer cells.Liu AX et al
    87302911996Familial cluster of ovarian small cell carcinoma: a new mendelian entity?Longy M et al
    94451311998Heterogeneous distribution of K-ras-mutated epithelia in mucinous ovarian tumors with special reference to histopathology.Mandai M et al
    20322351991Overexpression and mutation of p53 in epithelial ovarian cancer.Marks JR et al
    122182132002Characteristic pattern of genetic aberrations in ovarian granulosa cell tumors.Mayr D et al
    19235321991p53 mutations in ovarian cancer: a late event?Mazars R et al
    61477541984Comparative biochemical properties of normal and activated human ras p21 protein.McGrath JP et al
    85222481995DNA amplification of HER-2/neu and INT-2 oncogenes in epithelial ovarian cancer.Medl M et al
    114898162001Higher human kallikrein gene 4 (KLK4) expression indicates poor prognosis of ovarian cancer patients.Obiezu CV et al
    121491462002Allelotype analysis of common epithelial ovarian cancers with special reference to comparison between clear cell adenocarcinoma with other histological types.Okada S et al
    16805461991Frequent allelic losses and mutations of the p53 gene in human ovarian cancer.Okamoto A et al
    110166192000Identification by cDNA microarray of genes involved in ovarian carcinogenesis.Ono K et al
    81234691994Polymerase chain reaction allelotyping of human ovarian cancer.Osborne RJ et al
    109589372000Comparative genomic hybridization in inherited and sporadic ovarian tumors in Israel.Patael-Karasik Y et al
    77420041995Genetic changes in ovarian cancer.Pejovic T et al
    103855661999Well-differentiated mucinous carcinoma of the ovary and a coexisting Brenner tumor both exhibit amplification of 12q14-21 by comparative genomic hybridization.Pejovic T et al
    13770101992Chromosome aberrations in 35 primary ovarian carcinomas.Pejovic T et al
    85897231996Ovarian cancer risk in BRCA1 carriers is modified by the HRAS1 variable number of tandem repeat (VNTR) locus.Phelan CM et al
    85649791996Allelic deletion on chromosome 17p13.3 in early ovarian cancer.Phillips NJ et al
    23974551990Cytogenetic study of solid ovarian tumors.Roberts CG et al
    84947101993First experience with FGF-3 (INT-2) amplification in women with epithelial ovarian cancer.Rosen A et al
    95794281998BRCA1, BRCA2, and hereditary nonpolyposis colorectal cancer gene mutations in an unselected ovarian cancer population: relationship to family history and implications for genetic testing.Rubin SC et al
    13942081992Fine-scale deletion mapping of the distal long arm of chromosome 6 in 70 human ovarian cancers.Saito S et al
    19693811990Protooncogene amplification and tumor ploidy in human ovarian neoplasms.Sasano H et al
    13147751992Immunolocalization of c-myc oncoprotein in mucinous and serous adenocarcinomas of the ovary.Sasano H et al
    16552451991Allelotype of human ovarian cancer.Sato T et al
    84203531993Expression of ras oncogene p21 protein in normal and neoplastic ovarian tissues: correlation with histopathologic features and receptors for estrogen, progesterone, and epidermal growth factor.Scambia G et al
    91669521997Prognostic significance of ras/p21 alterations in human ovarian cancer.Scambia G et al
    22051001990C-myc proto-oncogene amplification detected by polymerase chain reaction in archival human ovarian carcinomas.Schreiber G et al
    117661422001Nongenetic screening of ovarian malignancies.Schwartz PE et al
    108530252000Detection of c-erbB-2 and FGF-3 (INT-2) gene amplification in epithelial ovarian cancer.Seki A et al
    114409952001Localization of a novel susceptibility gene for familial ovarian cancer to chromosome 3p22-p25.Sekine M et al
    118909882002Recurrent chromosome alterations in primary ovarian carcinoma in Chinese women.Sham JS et al
    99167991999PIK3CA is implicated as an oncogene in ovarian cancer.Shayesteh L et al
    76695551995The genetic analysis of ovarian cancer.Shelling AN et al
    96485891998No evidence for microsatellite instability from allelotype analysis of benign and low malignant potential ovarian neoplasms.Shih YC et al
    114792312001Genetic analysis of early- versus late-stage ovarian tumors.Shridhar V et al
    87005371996An abundance of p53 null mutations in ovarian carcinoma.Skilling JS et al
    24701521989Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer.Slamon DJ et al
    94087471997Comparative genomic hybridization detects frequent overrepresentation of chromosomal material from 3q26, 8q24, and 20q13 in human ovarian carcinomas.Sonoda G et al
    91159611997Detection of DNA gains and losses in primary endometrial carcinomas by comparative genomic hybridization.Sonoda G et al
    105779271999The genetic epidemiology of early-onset epithelial ovarian cancer: a population-based study.Stratton JF et al
    108950672000Genetic aberrations detected by comparative genomic hybridization in ovarian clear cell adenocarcinomas.Suehiro Y et al
    110340752000An approach to analysis of large-scale correlations between genome changes and clinical endpoints in ovarian cancer.Suzuki S et al
    103798761999Chromosome abnormalities in ovarian adenocarcinoma: I. Nonrandom chromosome abnormalities from 244 cases.Taetle R et al
    76067171995Mutation analysis of the BRCA1 gene in ovarian cancers.Takahashi H et al
    86328951996A 400 kb novel deletion unit centromeric to the BRCA1 gene in sporadic epithelial ovarian cancer.Tangir J et al
    94751951998C-myc mRNA expression in epithelial ovarian carcinomas in relation to estrogen receptor status, metastatic spread, survival time, FIGO stage, and histologic grade and type.Tanner B et al
    108159052000Frequent amplification of chromosomal region 20q12-q13 in ovarian cancer.Tanner MM et al
    91929821997Evidence for divergence of DNA copy number changes in serous, mucinous and endometrioid ovarian carcinomas.Tapper J et al
    114544212001Changes in gene expression during progression of ovarian carcinoma.Tapper J et al
    96618791998Genetic changes in inherited and sporadic ovarian carcinomas by comparative genomic hybridization: extensive similarity except for a difference at chromosome 2q24-q32.Tapper J et al
    86794431996Loss of heterozygosity on chromosome 5q in ovarian cancer is frequently accompanied by TP53 mutation and identifies a tumour suppressor gene locus at 5q13.1-21.Tavassoli M et al
    75456311995Abnormal expression of the retinoblastoma gene in ovarian neoplasms and correlation to p53 and K-ras mutations.Taylor RR et al
    83192181993p53 and Ki-ras gene mutations in epithelial ovarian neoplasms.Teneriello MG et al
    81740721994Clonal chromosome abnormalities in 54 cases of ovarian carcinoma.Thompson FH et al
    145539492003Genetic and cytogenetic observations among different types of ovarian tumors are compatible with a progression model underlying ovarian tumorigenesis.Tibiletti MG et al
    124076962002Cytogenetics and molecular genetics of ovarian cancer.Wang N et al
    113309452001A novel amplification at 17q21-23 in ovarian cancer cell lines detected by comparative genomic hybridization.Watanabe T et al
    145681722003Platelet-derived endothelial cell growth factor predicts of progression and recurrence in primary epithelial ovarian cancer.Watanabe Y et al
    96747051998Loss of heterozygosity on chromosomes 7p, 7q, 9p and 11q is an early event in ovarian tumorigenesis.Watson RH et al
    91909621997Loss of heterozygosity at the alpha-inhibin locus on chromosome 2q is not a feature of human granulosa cell tumors.Watson RH et al
    79592921994Molecular genetic changes associated with ovarian cancer.Weitzel JN et al
    93642941997Molecular genetics of gynecologic cancer.Whang JD et al
    90429081997Prevalence and contribution of BRCA1 mutations in breast cancer and ovarian cancer: results from three U.S. population-based case-control studies of ovarian cancer.Whittemore AS et al
    104731011999Decreased Src tyrosine kinase activity inhibits malignant human ovarian cancer tumor growth in a nude mouse model.Wiener JR et al
    115312842001Profiling of protein kinases in the neoplastic transformation of human ovarian surface epithelium.Wong AS et al
    97648301998Frequent loss of heterozygosity and three critical regions on the short arm of chromosome 8 in ovarian adenocarcinomas.Wright K et al
    80998991993Allelic loss in ovarian cancer.Yang-Feng TL et al
    98747981999NOEY2 (ARHI), an imprinted putative tumor suppressor gene in ovarian and breast carcinomas.Yu Y et al
    115981492001Comparative genomic hybridization of microdissected familial ovarian carcinoma: two deleted regions on chromosome 15q not previously identified in sporadic ovarian carcinoma.Zweemer RP et al
    77990451995Value of P-glycoprotein, glutathione S-transferase pi, c-erbB-2, and p53 as prognostic factors in ovarian carcinomas.van der Zee AG et al

    Citation

    Lisa Lee-Jones

    Ovary: Epithelial tumors

    Atlas Genet Cytogenet Oncol Haematol. 2003-12-01

    Online version: http://atlasgeneticsoncology.org/solid-tumor/5230/ovary-epithelial-tumors