Berek Novak's Gyn 2019. Chapter 39. Ovarian, Fallopian Tube, and Peritoneal Cancer

 Ovarian, Fallopian Tube, and Peritoneal Cancer

BS. Nguyen Hong Anh


KEY POINTS

1 The peak age incidence of invasive epithelial ovarian cancer is approximately 60

years. About 30% of ovarian neoplasms in postmenopausal women are malignant,

whereas only about 7% of ovarian epithelial tumors in premenopausal patients are

malignant. The average age at diagnosis of patients with borderline tumors is

approximately 46 years.

2 Ovarian cancer is associated with low parity and infertility. Because parity is

inversely related to the risk of ovarian cancer, having at least one child is protective,

with a risk reduction of 0.3 to 0.4.

3 Oral contraceptive use reduces the risk of epithelial ovarian cancer—women who use

oral contraceptives for 5 or more years reduce their relative risk by 0.5 (i.e., there is

a 50% reduction in the likelihood of development of ovarian cancer).

4 Given the false-positive results for CA-125 and transvaginal ultrasonography,

particularly in premenopausal women and the absence of evidence from randomized

trials that screening with CA-125 and TVUS reduces the mortality of ovarian

cancer, these tests should not be used routinely to screen women for ovarian cancer.

5 Most epithelial ovarian cancers are sporadic, but up to one-quarter of cases may be

associated with germline mutations in predisposing genes and may be hereditary.

Hereditary ovarian cancers, particularly those caused by BRCA1 mutations, occur at

a younger age, typically about 10 years earlier than sporadic ovarian cancers.

6 Most hereditary ovarian cancers result from germline mutations in the BRCA1 and

BRCA2 genes. The mutations are inherited in an autosomal dominant fashion, and

therefore a full pedigree analysis (i.e., maternal and paternal sides of the family

history for breast and ovarian cancer) must be carefully evaluated in all patients with

epithelial ovarian cancer, fallopian tube cancer, and peritoneal cancer. It is important

to note that almost 40% of women with BRCA-associated ovarian cancer do not

have a family history and hence genetic testing should be offered to all women with

ovarian cancer.

7 In unaffected women with a known BRCA1 mutation, the value of prophylactic

salpingo-oophorectomy is well established and is the most effective way to reduce

the risk of these cancers and reduce mortality.

8 The importance of thorough surgical staging cannot be overemphasized, because

subsequent treatment and prognosis will be determined by the stage of the disease.

All patients with suspected ovarian cancer should be evaluated by a gynecologic

oncologist. Patients with advanced-stage ovarian cancer (stage IIIC or IV) should

have clinical evaluation including chest, abdomen and pelvis imaging. Patients

determined to have a poor performance status, a high perioperative risk, or a low

probability of attaining optimal cytoreduction to no visible, or palpable, residual

2543disease, or at least cytoreduction to less than 1 cm of residual disease, should be

considered for neoadjuvant chemotherapy prior to attempt at interval cytoreductive

surgery after two to three cycles of chemotherapy. Patients with advanced ovarian

cancer who are medically fit for major surgery and who potentially have resectable

disease should be offered primary cytoreductive surgery followed by adjuvant

chemotherapy. In the United States, primary cytoreductive surgery is preferred, if

there is a high likelihood of achieving optimal cytoreduction with acceptable

morbidity from surgery. Prior to initiating neoadjuvant chemotherapy, the

pathologic diagnosis of ovarian, fallopian tube, or peritoneal cancer must be made

on biopsy.

9 Combination chemotherapy with carboplatin and paclitaxel (CP) is recommended for

patients with high-risk, low-stage disease. For advanced-stage epithelial ovarian

cancer, the choice of intravenous (IV) versus intraperitoneal (IP) platinum and

taxane chemotherapy with the addition of bevacizumab should be individualized.

10 The ICON8 trial did not confirm the JGOG study that showed that a dose-dense

regimen of weekly paclitaxel and 3-weekly carboplatin was significantly superior to

the standard every 3-week chemotherapy regimen of both drugs. Therefore 3-weekly

regimens remain the standard of care in non-Japanese women.

11 BRCA-associated ovarian cancers have increased sensitivity to platinum

chemotherapy and to poly (ADP-ribose) polymerase (PARP) inhibitors. PARP

inhibitors are approved to treat selected patients with recurrent ovarian cancer and

for maintenance therapy following response to platinum-based chemotherapy in the

recurrent setting. There is good evidence from a number of randomized trials that

maintenance therapy with PARP inhibitors results in a significant increase in the

progression-free survival (PFS), particularly in BRCA mutation carriers (including

tumors with somatic BRCA mutations). PARP inhibitors niraparib and olaparib are

U.S. Food and Drug Administration (FDA) approved for maintenance therapy in

patients with platinum-sensitive disease with a partial or complete response to

platinum-based chemotherapy. PFS is improved with these agents regardless of

BRCA status, although the PFS is significantly longer in patients with BRCA

mutations. Maintenance therapy with rucaparib was reported to be associated with

significant improvement in PFS in patients with platinum-sensitive recurrent ovarian

cancer after response to chemotherapy, with the benefits being greater in women

with somatic or germline BRCA (gBRCA) mutations.

12 In the first two decades of life, almost 70% of ovarian tumors are of germ cell

origin, and one-third of these are malignant. In contrast to the relatively slowgrowing epithelial ovarian tumors, germ cell malignancies grow rapidly.

13 The most common types of malignant germ cell tumors are dysgerminomas,

immature teratomas, and endodermal sinus tumors (EST). Preservation of fertility

should be standard in most patients. The most effective chemotherapy is bleomycin,

etoposide, and cisplatin (BEP).

14 Stromal tumors include adult granulosa cell tumors, which are low-grade

malignancies. They account for 70% of ovarian sex cord–stromal tumors. Granulosa

2544cell tumors are the most common type of malignant ovarian sex cord–stromal

tumors. In premenopausal women, they can be treated conservatively. Adjuvant

chemotherapy is of unproven value.

15 Metastatic tumors to the ovaries are most frequently from the breast and

gastrointestinal tract.

16 Epithelial serous fallopian tube carcinomas and peritoneal cancers are the same as

ovarian cancer and are treated in the same way, with staging and cytoreductive

surgery followed by platinum and taxane chemotherapy. The distinction between

high-grade serous ovarian, fallopian tube, and peritoneum cancers is an anachronism

because as most high-grade serous cancers arise from the fimbriae of the fallopian

tube.

Ovarian cancer is the second most common gynecologic cancer in the United

States but it accounts for the majority of deaths from these cancers. Epithelial

cancers are the most common ovarian/fallopian malignancy, and at initial

diagnosis, over two-thirds of patients have advanced disease. Ovarian cancer

represents a major surgical challenge as the aim is to resect all visible disease.

Optimal therapy includes maximal attempt at surgical cytoreduction to no gross

disease, or at least optimal debulking (to <1 cm of residual disease), followed by

platinum-based combination chemotherapy. It has the highest fatality-to-case ratio

of all the gynecologic malignancies. There are nearly 22,280 new cases annually

in the United States, and 14,240 women can be expected to succumb to their

illness (1). Ovarian cancer is the seventh most common cancer in women in the

United States, accounting for 3% of all malignancies, 6% of deaths from cancer in

women, and almost one-third of invasive malignancies of the female genital

organs. Ovarian cancer is the fifth most common cause of death from malignancy

in women. The lifetime risk of being diagnosed with ovarian cancer is 1% to

1.5% and of dying from ovarian cancer is almost 0.5% (2).

EPITHELIAL OVARIAN CANCER

Developments, based in part on meticulous pathologic assessment of riskreducing salpingo-oophorectomy specimens from BRCA patients, have

completely changed our perception and understanding of “ovarian” cancer.

While previously thought to be derived from the coelomic epithelium on the

surface of the ovary or from ovarian inclusion cysts, it is now apparent that

most serous carcinomas originate from the fallopian tube (3), while other

subtypes (clear cell, endometrioid) are derived from endometriosis. A detailed

classification of the histologic types of epithelial tumors of the ovary is presented

in Table 39-1. Neoplastic transformation can occur when the cells are genetically

2545predisposed to oncogenesis, exposed to an oncogenic agent, or both (4).

Table 39-1 Epithelial Ovarian Tumors

Histologic Type Cellular Type

I. Serous Endosalpingeal

A. Benign

B. Borderline

C. Malignant

II. Mucinous Intestinal, endocervical

A. Benign

B. Borderline

C. Malignant

III. Endometrioid Endometrial

A. Benign

B. Borderline

C. Malignant

IV. Clear-cell Müllerian

A. Benign

B. Borderline

C. Malignant

V. Brenner Transitional

A. Benign

B. Borderline (proliferating)

C. Malignant

VI. Mixed epithelial Mixed

2546A. Benign

B. Borderline

C. Malignant

VII. Undifferentiated May be anaplastic

VIII. Unclassified

From Seroy SF, Scully RE, Sobin LH. International Histological Classification of

Tumours No. 9. Histological Typing of Ovarian Tumors. Geneva, Switzerland: World

Health Organization; 1973, with permission.

Pathology

Invasive Cancer

Seventy-five percent to 80% of epithelial cancers are of the serous histologic

type. Less common types are endometrioid (10%), clear cell (5%), mucinous

(5%), transitional, and undifferentiated carcinomas, with the last two types

representing less than 1% of epithelial lesions (2). Each of the major tumor

types is named on the basis of a histologic pattern that resembles the epithelium in

the lower genital tract (4). For example, serous tumors have an appearance similar

to that of the glandular epithelial lining of the fallopian tube, endometrioid tumors

resemble proliferative endometrium, and clear cell tumors resemble secretory or

gestational endometrium. Mucinous tumors may contain cells that resemble

endocervical glands, but more commonly these cells resemble the gastrointestinal

epithelium. Transitional tumors are so named because of a resemblance to the

epithelium in Walthard rests and bladder urothelium.

There is growing evidence that many, if not most, high-grade serous

carcinomas of the ovary arise from the fimbrial end of the fallopian tube rather

than from the ovary (5,6). Serous epithelial ovarian cancers can be separated

into two distinct groups—type I and type II serous tumors—as they differ

considerably in the cell of origin, molecular pathogenesis, and their biologic

behavior (7). Type I tumors include serous borderline tumors and low-grade

serous carcinoma; they are genetically stable, do not have p53 mutations,

and may have mutations in KRAS or BRAF. In contrast, type II serous

tumors are rapidly growing, highly aggressive neoplasms that may have

precursor lesions in the fimbriae of the fallopian tube; most are advanced

stage at diagnosis (8). The type II tumors are genetically unstable and almost

universally have p53 mutations.

2547Borderline Tumors

An important group of tumors to distinguish are tumors of low malignant

potential, also called the borderline tumors. Borderline tumors tend to remain

confined to the ovary for long periods, they occur predominantly in

premenopausal women, and are associated with a very good prognosis

(2,4,8–13). They are diagnosed most frequently in women between the ages of 30

and 50 years, whereas invasive carcinomas occur more often in women between

the ages of 50 and 70 years (2).

Although uncommon, extraovarian implants may occur with serous

borderline tumors. These implants are characterized as either noninvasive or

invasive (analogous to low-grade serous carcinoma). The latter has a higher

likelihood of progression, in the peritoneal cavity, which can ultimately lead to

intestinal obstruction caused by associated fibrosis and death (2,7).

Classification of Epithelial Ovarian Tumors

Serous Tumors

Serous tumors are so classified because they resemble tubal secretory cells.

Psammoma bodies are frequently found in these neoplasms, and they are made

up of concentric rings of calcification. Several hypotheses pertaining to the origin

and development of psammoma bodies are proposed, including apoptosis of

tumor cells and osteoinductive cytokines produced by macrophages, which result

in calcification that can be extensive (7). In the wall of the mesothelial

invaginations, papillary ingrowths are common, representing the early stages of

development of a papillary serous cystadenoma. There are many variations in the

proliferation of these mesothelial inclusions. Several foci may be lined with

flattened inactive epithelium; in adjacent cavities, papillary excrescences are

present, often resulting from local irritants (2).


FIGURE 39-1 Serous borderline tumor of the ovary. Complex papillary fronds with

hierarchical branching are lined with pseudostratified columnar cells. The epithelium and

the stroma are clearly separated by a basement membrane, indicating no stromal invasion.

Borderline Serous Tumors

Approximately 10% of all ovarian serous tumors fall into the category of a

tumor of low malignant potential or borderline tumor (Fig. 39-1), and 50%

occur before the age of 40 years. The criteria for the diagnosis of serous

borderline tumors are as follows (12):

1. Epithelial hyperplasia in the form of pseudostratification, tufting, cribriform,

and micropapillary architecture

2. Mild nuclear atypia and mild increased mitotic activity

3. Detached cell clusters

4. Absence of destructive stromal invasion (i.e., without tissue destruction)

Serous borderline tumors that are composed of an exuberant micropapillary

architecture are designated as serous borderline tumors with micropapillary

2549features (Fig. 39-2); these tumors are more frequently bilateral, exophytic, and

high stage than the usual serous borderline tumor.

It should be emphasized that up to 40% of serous borderline tumors are

associated with spread beyond the ovary, but high-stage disease does not

necessarily warrant a diagnosis of an invasive carcinoma. The diagnosis of a

serous borderline tumor versus serous carcinoma is based on the histologic

features of the primary tumor (12). Up to 10% of women with ovarian serous

borderline tumors and extraovarian implants may have invasive implants (i.e.,

low-grade serous carcinoma), and these can behave more aggressively (14). The

5-year overall survival for women with invasive implants (low-grade serous

carcinoma) is about 50% if stringent criteria are applied (11,14–16). Most

implants are noninvasive (11,17). In the noninvasive implants, papillary

proliferations of atypical cells involve the peritoneal surface and form smooth

invaginations (11). In contrast, the invasive implants resemble well-differentiated

serous carcinoma and are characterized by atypical cells forming irregular glands

with sharp borders. Implants are usually confined to the abdominal cavity and

may be seen in the pelvis, omentum, and adjacent tissues, including lymph nodes,

but spread outside the abdominal cavity is rare. Death can occur as the result of

intestinal obstruction (17–20).

Borderline serous tumors may harbor foci of stromal microinvasion (19).

Most patients are young and their tumors are International Federation of

Obstetrics and Gynecology (FIGO) stage I. Stromal microinvasion is increased

about ninefold in pregnant women with serous borderline tumors.

The presence of stromal microinvasion is associated with lymphovascular

space invasion in the primary ovarian tumor (and likely represents a form of true

stromal invasion), but it is not associated with an aggressive clinical course, and

patients with this finding should be managed in the same way as patients without

stromal microinvasion.

Serous Carcinomas

In malignant serous tumors, stromal invasion is present (2). The grade of

tumor is important and needs to be documented. In low-grade serous

adenocarcinomas, papillary and glandular structures predominate (Fig. 39-3);

high-grade neoplasms are characterized by solid sheets of cells, nuclear

pleomorphism, and high mitotic activity (Fig. 39-4). Laminated, calcified

psammoma bodies are found in 80% of serous carcinomas. Serous

psammocarcinoma is a rare variant of serous carcinoma characterized by

massive psammoma body formation and low-grade cytologic features. At

least 75% of the epithelial nests are associated with psammoma body formation.

Patients with serous psammocarcinoma have a protracted clinical course and a

2550relatively favorable prognosis; their clinical course more closely resembles that of

high-stage, progressive serous borderline tumor than serous carcinoma.

FIGURE 39-2 Serous borderline tumor with micropapillary features. The papillae have a

nonhierarchical branching pattern and are lined by a monomorphous population of cells.

Mucinous Tumors

These cystic ovarian tumors have loculi lined with mucin-secreting epithelium.

The lining epithelial cells contain intracytoplasmic mucin and resemble those of

endocervix, gastric pylorus, or intestine. They represent about 8% to 10% of

epithelial ovarian tumors. They may reach enormous size, filling the entire

abdominal cavity (2).


FIGURE 39-3 Low-grade serous adenocarcinoma of the ovary. Clusters and papillae of

malignant cells are in direct contact with fibrous stroma indicative of stromal invasion.

Borderline Mucinous Tumors

The mucinous tumor of low malignant potential is often difficult to diagnose.

Although it is common to find a rather uniform pattern from section to section in

the serous borderline tumor, this is not true in the mucinous tumors. Welldifferentiated mucinous epithelium may be seen immediately adjacent to a

poorly differentiated focus. It is important to take multiple sections from

many areas in the mucinous tumor to identify the most malignant alteration

(2).


FIGURE 39-4 High-grade serous adenocarcinoma. Papillae lined by sheets of

cytologically malignant cells invade stroma, often with associated necrosis.


FIGURE 39-5 Mucinous adenocarcinoma of the ovary. Irregular glandular spaces are

lined with a layer of tall columnar cells with abundant mucinous cytoplasm, resembling

intestinal epithelium at the left. At the right, there is destructive invasion into the ovarian

stroma.

Mucinous Carcinomas

Bilateral tumors occur in 8% to 10% of cases. The mucinous lesions are confined

to the ovary in 95% to 98% of cases (Fig. 39-5). Because most ovarian mucinous

carcinomas contain enteric-type cells, they cannot be distinguished from

metastatic carcinoma of the gastrointestinal tract on the basis of histology alone.

Primary ovarian neoplasms rarely metastasize to the mucosa of the bowel,

although they commonly involve the serosa, whereas gastrointestinal lesions

frequently involve the ovary by direct extension or lymphatic spread (2).

Pseudomyxoma Peritonei

Pseudomyxoma peritonei is a clinical term used to describe the finding of

abundant mucoid or gelatinous material in the pelvis and abdominal cavity

surrounded by fibrous tissue. It is most commonly secondary to a well-

2554differentiated appendiceal mucinous neoplasm or other gastrointestinal

primary; rarely, mucinous tumors arising in an ovarian mature teratoma are

associated with pseudomyxoma peritonei.

Endometrioid Tumors

Endometrioid lesions constitute 6% to 8% of epithelial tumors. Endometrioid

neoplasia includes all the benign demonstrations of endometriosis. In 1925,

Sampson suggested that certain cases of adenocarcinoma of the ovary probably

arose in areas of endometriosis (20). The adenocarcinomas are similar to those

seen in the uterine corpus. The malignant potential of endometriosis is very low,

although a transition from benign to malignant epithelium may be demonstrated.

Borderline Endometrioid Tumors

The endometrioid tumor of low malignant potential has a wide morphologic

spectrum. Tumors may resemble an endometrial polyp or complex endometrial

hyperplasia with glandular crowding. When there are back-to-back,

architecturally complex glands with no intervening stroma, the tumor is classified

as a well-differentiated endometrioid carcinoma. Some borderline endometrioid

tumors have a prominent fibromatous component. In such cases, the term

adenofibroma is used to describe them (2).

Endometrioid Carcinomas

Endometrioid tumors are characterized by a markedly complex glandular pattern

with all the potential variations of epithelia found in the uterus (Fig. 39-6).


FIGURE 39-6 Endometrioid cancer. Round to tubular glands lined by stratified columnar

cells with a confluent growth pattern.

Multifocal Disease

The endometrioid tumors afford the greatest opportunity to evaluate multifocal

disease. Endometrioid carcinoma of the ovary is associated in 15% to 20% of

the cases with carcinoma of the endometrium. Identification of multifocal

disease is important because patients with disease metastatic from the uterus

to the ovaries have a 30% to 40% 5-year survival, whereas those with

synchronous multifocal disease have a 75% to 80% 5-year survival (21).

When the histologic appearance of endometrial and ovarian tumors is different,

the two tumors most likely represent two separate primary lesions. When they

appear similar, the endometrial tumor can be considered a separate primary tumor

if it is well differentiated and only superficially invasive.

Clear Cell Carcinomas

Several basic histologic patterns are present in the clear cell adenocarcinoma (i.e.,

tubulocystic, papillary, reticular, and solid). The tumors are made up of clear and

2556hobnail cells that project their nuclei into the apical cytoplasm. The clear cells

have abundant clear or vacuolated cytoplasm, hyperchromatic irregular nuclei,

and nucleoli of various sizes (Fig. 39-7). Focal areas of endometriosis are

common and mixed clear cell and endometrioid carcinoma may occur (21).

The clear cell carcinoma seen in the ovary is histologically identical to that seen

in the uterus or vagina of the young patient who has been exposed to

diethylstilbestrol (DES) in utero. Almost invariably high-grade nuclei are

identified. Hence, clear cell carcinomas are not graded.

Transitional (Brenner) Tumors

Borderline Brenner Tumors

Proliferative Brenner tumors were previously subclassified as proliferating tumors

(those tumors that resemble low-grade papillary urothelial carcinoma of the

urinary bladder) and borderline tumors (those tumors that resemble high-grade

papillary urothelial carcinoma), these groups of tumors are now classified as

borderline Brenner tumors (22). Complete surgical removal usually results in

cure.


FIGURE 39-7 Clear cell carcinoma of the ovary. Note the solid variant of clear cell

carcinoma with sheets of cells that have clear cytoplasm (“hobnail” cells).

Malignant Brenner Tumors

These rare tumors are defined as benign or borderline Brenner tumors

coexisting with invasive transitional cell carcinoma.

Transitional Cell Carcinoma

The designation transitional cell carcinoma has been attributed to primary

ovarian carcinoma resembling transitional cell carcinoma of the urinary

bladder without a recognizable Brenner tumor. It is now thought that these are

a variant of high-grade serous carcinoma and do not represent a distinct epithelial

subtype of carcinoma (23,24). Unlike malignant Brenner tumors these tumors are

more frequently diagnosed in an advanced stage and are associated with a

survival rate similar to that of high-grade serous carcinoma (25).

Peritoneal Carcinomas

Peritoneal tumors are histologically indistinguishable from ovarian and

fallopian tube serous tumors. In the case of borderline serous peritoneal

tumors and serous peritoneal carcinomas, the ovaries are normal or minimally

involved, and the tumors affect predominantly the uterosacral ligaments, pelvic

peritoneum, or omentum. The overall prognosis for borderline serous peritoneal

tumors is excellent and comparable to that of ovarian borderline serous tumors

(26–28). In the review of 38 cases of peritoneal borderline serous tumors from the

literature, 32 women had no persistent disease, 4 were well after resection of

recurrence, 1 developed an invasive serous carcinoma, and 1 died from the effects

of the tumor (26).

Carcinomas that appear to be predominantly peritoneal without

appreciable ovarian or fallopian tube enlargement have traditionally been

called peritoneal carcinoma, when tumors spread from the breast,

gastrointestinal tract, and other organs of non-müllerian origin are excluded.

Most are peritoneal serous carcinomas, and have the appearance of a moderately

to poorly differentiated serous ovarian carcinoma. Peritoneal endometrioid

carcinoma also occurs, but is less common; peritoneal clear cell carcinoma is rare.

[15] Peritoneal serous carcinoma should be considered clinically the same as

ovarian and fallopian tube cancers, as all these cancers share a similar

pathogenesis and clinical behavior and treatment. Patients in whom

exploratory surgery is performed, there may be microscopic or small macroscopic

cancer on the surface of the ovary and extensive disease in the upper abdomen,

particularly in the omentum (29).

2558Mesotheliomas

Peritoneal malignant mesotheliomas may be epithelial, sarcomatous, or

biphasic (2,30). Deciduoid peritoneal mesothelioma is an unusual variant that

resembles exuberant, ectopic decidual reaction of the peritoneum. Asbestos

exposure is not correlated with peritoneal mesotheliomas in women. These

lesions typically appear as multiple intraperitoneal (IP) masses, often coating the

entire peritoneum and can develop after hysterectomy and bilateral salpingooophorectomy for benign disease. Malignant mesotheliomas should be

distinguished from benign multicystic peritoneal mesothelioma (multilocular

peritoneal inclusion cyst), and ovarian tumor implants and primary peritoneal

müllerian neoplasms.


FIGURE 39-8 Ovarian cancer incidence: distribution by age. (From Nagy K. The side

effects of managed care on the drug industry. J Natl Cancer Inst 1995;87:1280, with

2559permission.)

Clinical Features

More than 80% of epithelial ovarian cancers are found in postmenopausal women

(Fig. 39-8). The peak incidence of invasive epithelial ovarian cancer is at 56

to 60 years of age (2,3). The age-specific incidence of ovarian epithelial cancer

rises precipitously from 20 to 80 years of age and subsequently declines. These

cancers are relatively uncommon in women younger than age 45. Fewer than 1%

of epithelial ovarian cancers occur before the age of 21 years, two-thirds of

ovarian malignancies in such patients being germ cell tumors (2,31). [1] About

30% of ovarian neoplasms in women are malignant, whereas only about 7%

of ovarian epithelial tumors in premenopausal patients are malignant (2,3).

The average age of patients with borderline tumors is approximately 46

years (2,3,9). Eighty percent to 90% of ovarian cancers, including borderline

forms, occur after the age of 40 years, whereas 30% to 40% of malignancies

occur after the age of 65 years. The chance that a primary epithelial tumor will be

of borderline or invasive malignancy in a patient younger than 40 years is

approximately 1 in 10, but after that age it rises to 1 in 3 (2,3). Less than 1% of

epithelial ovarian cancers occur before the age of 20 years, with two-thirds of

ovarian malignancies in such patients being germ cell tumors (31).

Etiology

[2] Ovarian cancer is associated with low parity and infertility (32). Although

there are a variety of epidemiologic variables correlated with ovarian cancer, such

as talc use, galactose consumption, and tubal ligation, none is as strongly

correlated as prior reproductive history and duration of the reproductive career

(32,33). Early menarche and late menopause increase the risk of ovarian cancer

(33). These factors and the relationship of parity and infertility to the risk of

ovarian cancer led to the hypothesis that suppression of ovulation may be an

important factor.

Prevention

[2] Because parity is inversely related to the risk of ovarian cancer, having at

least one child is protective for the disease, with a risk reduction of 0.3 to 0.4.

Oral contraceptive use reduces the risk of epithelial ovarian cancer [3] (32).

Women who use oral contraceptives for 5 or more years reduce their relative

risk to 0.5 (i.e., there is a 50% reduction in the likelihood of development of

ovarian cancer). Women who had two children and used oral contraceptives for

5 or more years have a relative risk of ovarian cancer as low as 0.3, or a 70%

2560reduction (34). The oral contraceptive pill is the only documented method of

chemoprevention for ovarian cancer, and it should be recommended to

women for this purpose. When counseling patients regarding birth control

options, this important benefit of oral contraceptive use should be

emphasized. This is important for women with a strong family history of

ovarian cancer.

The performance of a prophylactic salpingo-oophorectomy significantly

reduces, but does not totally eliminate, the risk of nonuterine pelvic cancers;

because the entire peritoneum is at risk, peritoneal carcinomas can occur in 2% to

3% of women even after prophylactic bilateral salpingo-oophorectomy (26,29).

A thorough discussion of the risks and benefits of oophorectomy should be

undertaken in premenopausal women who are undergoing a hysterectomy

for benign disease, do not carry germline mutations, and do not have a

family history that suggests that they are at higher than average risk for

ovarian cancer (35). The ovaries may provide protection from cardiovascular

disease and osteoporosis, and long-term mortality may not be decreased by

the performance of prophylactic oophorectomy in women at population risk

of ovarian cancer (36).

Screening

CA-125 is useful for monitoring epithelial ovarian cancer patients during their

chemotherapy; and is elevated in 50% of patients with stage I disease and in 80%

to 90% of patients with advanced serous cancers (37–46). Transvaginal

ultrasonography is useful to characterize the morphology of ovarian tumors

(47,48). However, the value of tumor markers and ultrasonography to screen

for epithelial ovarian cancer is not established by prospective studies (49). [4]

The use of transvaginal ultrasound alone or in combination with CA-125 for

early ovarian cancer detection has not proven effective in average-risk

women to decrease mortality from ovarian cancer, instead there are harms

to this strategy, including false-positive results and more invasive procedures

being performed such as surgery (50). The inability to use these modalities for

screening is discussed in the American College of Obstetricians and

Gynecologists (ACOG) committee opinion (51).

The U.S. Preventative Services Task Force (USPSTF) recommends against

screening asymptomatic women for ovarian cancer with pelvic examination,

pelvic ultrasound, or serum tumor marker measurements (52). The USPSTF

concluded that there is adequate evidence that there is no mortality benefit to

routine screening for ovarian cancer with transvaginal ultrasonography or

single-threshold serum CA-125 testing and that the harms of such screening

are at least moderate. There is no effective strategy for early detection of

2561ovarian cancer that translates to a reduction in ovarian cancer mortality

(52). Ultrasonography, despite being able to detect changes in size and

morphology of the ovaries, has not been shown be effective for ovarian

cancer screening (2,5).

Final results from United Kingdom Collaborative Trial of Ovarian Cancer

Screening (UKCTOCS), which is fully accrued and follow-up is pending,

should provide more information about the relative benefits and harms of an

algorithm-based approach to screening for ovarian cancer. In the UKCTOCS

study, 202,638 healthy average-risk postmenopausal patients in the age range 50

to 74 years were randomized to receive no screening, annual pelvic ultrasound or

multimodal screening using a combination of CA-125 testing (interpreted using

risk of ovarian cancer algorithm [ROCA]) and ultrasound triggered by a ROCA

score signifying increased risk. Multimodal screening with the ROCA was

superior to using fixed CA-125 cut-off values in combination with ultrasound

screening (positive predictive value = 35.1%). Ten women had surgical

intervention because of a false-positive screening result, demonstrating that the

harm related to unnecessary surgical procedures as a consequence of a falsepositive test can occur (50,53). Thus far, follow-up data has not shown a

significant reduction of ovarian cancer mortality with multimodal screening when

compared with no screening (50,54).

The ROCA uses serially measured CA-125 and mathematical modeling to

determine whether an increase in levels should prompt referral for ultrasound and

has a better predictive performance for detecting ovarian cancer than using a fixed

CA-125 value cut-off (55,56–59). The U.S. Food and Drug Administration (FDA)

has warned that early detection tests, including the ROCA test, are unreliable

(60).

Other large prospective randomized trials in the United States and the United

Kingdom sought to address the utility of a multimodality assessment for early

detection of ovarian cancer. In the US-based Prostate, Lung, Colorectal, and

Ovarian (PLOC) Cancer Screening Trial, 78,216 healthy average-risk women

between age 55 and 74 years were randomized to have either annual CA-125

testing for 6 years and transvaginal ultrasound examination for 4 years (tested

group) versus the “usual care” who were patients not offered this testing, many of

these patients did have annual pelvic examinations. There was an ovarian cancer

mortality rate of 3.1 deaths per 10,000 person-years versus 2.6 deaths per 10,000

person-years in the tested group versus usual care group respectively, after a

median follow-up period of 12.4 years. This study also showed that using a

baseline and annual measurement of CA-125 with a cut-off (35 U/mL) in

combination with pelvic ultrasound as a way for early assessment does not

result in decreased ovarian cancer mortality (45,61). Thus far, this large trial

2562showed that screening women at average risk with annual transvaginal

ultrasound provided no mortality benefit when compared with no screening.

In the future, new markers or technologies may improve the specificity of

ovarian cancer screening, but proof of this will require a large, prospective study

(41,42).

Screening in women who have a familial risk may provide a better yield, but

there is no evidence to demonstrate a benefit of screening high-risk women,

even with active investigation (62,63). The findings of two prospective studies of

annual transvaginal ultrasound and CA-125 screening in 888 BRCA1 and BRCA2

mutation carriers in the Netherlands and 279 mutation carriers in the United

Kingdom are not encouraging and suggest a very limited benefit of screening in

high-risk women (62,64). Despite annual gynecologic screening, Hermsen et al.

reported that a high proportion of ovarian cancers in BRCA1 and BRCA2 carriers

were interval cancers and the large majority of all cancers diagnosed were at

advanced stages; similar results were reported by Woodward et al. (62,64).

Women with particular genetic syndromes are at increased risk of ovarian

cancer, for example patients with a BRCA gene mutation or Lynch syndrome

(hereditary nonpolyposis colorectal cancer). These patients should be referred to

genetic counseling to have an evaluation in order to properly quantify their level

of risk and have a discussion about the available risk reduction strategies.

Genetic Risk for Epithelial Ovarian Cancer

The lifetime risk of ovarian carcinoma for women in the United States is about

1.4% (2,4,65). The risk of ovarian cancer is higher than that in the general

population in women with certain family histories (55,62–64,66–71). [5] Most

epithelial ovarian cancer is sporadic, with causes accounting for up to 24%

of invasive epithelial ovarian cancer (14).

Hereditary Ovarian Cancer

BRCA1 and BRCA2

Most hereditary ovarian cancers result from pathogenic mutations in either

the BRCA1 or BRCA2 genes. Many studies have found that at least 15% of

women with high-grade nonmucinous ovarian cancers have germline

mutations in BRCA1/BRCA2 and almost 40% of these women do not have a

family history of breast/ovarian cancer (71). There are other low-to-moderate–

penetrance genes that predispose to ovarian cancer, and this is an area of intense

research interest (65,72,73). Norquist et al. used next-generation sequencing of

constitutional DNA samples from over 1,900 women with ovarian cancer to

identify germline mutations in a panel of 20 genes, including BRCA1 and BRCA2,

2563DNA mismatch repair genes, double-stranded DNA break repair genes such as

CHEK2 and ATM, and the BRCA1-associated complex or the BRCA2/Fanconi

Anemia pathway genes (including BRIP1, BARD1, PALB2, RAD50, RAD51C,

RAD51D among others). About 80% of mutations were in BRCA1 or BRCA2.

Approximately 3% of patients carried mutations in the Fanconi Anemia pathway

genes, while only 0.4% had mutations in mismatch repair genes (74).

[6] The mutations are inherited in an autosomal dominant fashion, and

therefore a full pedigree analysis (i.e., maternal and paternal sides of the

family) is essential when taking a family history. All women with high-grade

invasive ovarian cancers should be offered genetic testing, even if they do not

have a family history of breast/ovarian cancer (75). A very large study that

followed women enrolled in familial cancer registries primarily in the United

Kingdom, Holland, and France provides the most comprehensive data on the risk

of ovarian cancer and breast cancer in BRCA mutation carriers. Women were

recruited into this cohort from 1997 to 2011 and had not undergone risk-reducing

breast or gynecologic surgery; follow-up continued until 2013 (median, 5 years).

A total of 6,036 BRCA1 and 3,820 BRCA2 mutation carriers were followed

(median age at study entry, 38). The cumulative risk for breast cancer by age 80

was 72% in BRCA1 carriers and 69% in BRCA2 carriers. For ovarian cancer, the

cumulative risk was 44% and 17%, respectively. The peak breast cancer incidence

occurred at age 41 to 50 for BRCA1 carriers and age 51 to 60 for BRCA2 carriers.

The incidence of ovarian cancer was 3.6 times higher among BRCA1 than BRCA2

carriers.

[5] Hereditary ovarian cancers occur in women who are approximately 10

years younger than those with nonhereditary tumors (i.e., closer to age 50

compared to age 60 for those with sporadic cancer) (70). A woman with a first- or

second-degree relative who had premenopausal ovarian cancer may have a higher

probability of being a BRCA carrier. Women with BRCA germline mutations

tend to be diagnosed at a younger age than women with sporadic ovarian cancers.

In women with BRCA1 mutations, less than 2% to 3% of carriers develop ovarian

cancer by the age of 40 which increases to 10% to 21% by the age of 50. In

women with BRCA2 mutations, less than 3% of carriers develop ovarian cancer

by the age of 50 and most develop after age 50. Most BRCA ovarian cancers are

high-grade serous carcinomas (Fig. 39-9).


FIGURE 39-9 BRCA1-associated ovarian carcinoma is typically a high-grade serous

adenocarcinoma with numerous mitotic figures and marked nuclear pleomorphism. A brisk

lymphocytic infiltrate with tumor-infiltrating lymphocytes is not uncommon in these

tumors.

Founder Effect

There is a higher carrier rate of BRCA1 and BRCA2 mutations in women of

Ashkenazi Jewish descent, Icelandic women, and many other ethnic groups

(76–81). There are three specific founder mutations carried by the Ashkenazi

population, 185delAG and 5382insC on BRCA1, and 6174delT on BRCA2.

Individuals of Ashkenazi Jewish descent have a 1 in 40, or 2.5%, chance of

having a mutation in BRCA1 or BRCA2, in contrast to a 1 in 500 risk in the

Caucasian population (82). The increased risk is a result of the founder effect, in

which a higher rate of specific mutations occurs in an ethnic group from a defined

geographic area. These founder mutations generated considerable interest,

because they facilitate studies of prevalence and penetrance and can be used to

quantify the degree of homogeneity within a population.

2565Pedigree Analysis

[6] The risk of carrying a germline mutation that predisposes to ovarian

cancer depends on the number of first- or second-degree relatives (or both)

with a history of epithelial ovarian carcinoma or breast cancer (or both) and

on the number of malignancies that occur at an earlier age. The degree of

risk is difficult to determine precisely without performing a full pedigree

analysis. It is important to be aware that 40% of women with BRCA-related

ovarian cancer do not have a family history.

Lynch Syndrome or Hereditary Nonpolyposis Colon Cancer

Lynch syndrome (HNPCC), which includes multiple adenocarcinomas,

involves a combination of colon cancer and endometrial or ovarian cancer

and other malignancies of the gastrointestinal and genitourinary systems

(83). The chief gene mutations that are associated with this syndrome include

MSH2, MLH1, MSH6, and PMS2 or the EPCAM gene. The risk that a woman

who is a member of one of these families will develop epithelial ovarian cancer

depends on the frequency of this disease in first- and second-degree relatives,

although these women appear to have at least three times the relative risk of the

general population. A full pedigree analysis of such families should be performed

by a geneticist to more accurately determine the risk.

Management of Women at High Risk for Ovarian Cancer

The management of an unaffected woman with a germline BRCA (gBRCA)

mutation or a strong family history of epithelial ovarian cancer where a

mutation has not been found must be individualized and depends on her age,

reproductive plans, and the estimated risk. All these women should be seen by

a geneticist in a high-risk clinic. Decisions about management are best made after

careful study and, whenever possible, verification of the histologic diagnosis of

the family members’ ovarian cancer.

The value of testing for BRCA1 and BRCA2 is well established, and there

are guidelines for testing (75,82,84,85). Essentially all women with epithelial

ovarian cancer, excluding mucinous cancers, should be offered testing

irrespective of whether she has a family history of breast/ovarian cancer. The

American Society of Clinical Oncology offered guidelines that emphasize careful

evaluation by geneticists, careful maintenance of medical records, and a genetic

screening clinic with an understanding of how to effectively counsel and manage

these patients. Concerns remain over the use of the information, impact on

insurability, interpretation of the results, and how the information will be used

within a specific family (e.g., to counsel children).

Although there are some conflicting data, the behavior of breast cancers arising

2566in women with germline mutations in BRCA1 or BRCA2 is comparable to the

behavior of sporadic tumors (86,87). Women with breast cancer who carry

these mutations are at a greatly increased risk of ovarian cancer and a

second breast cancer: the lifetime risk of ovarian cancer is 54% for women

who have a BRCA1 mutation and 23% for those with a BRCA2 mutation,

and for the two groups together, there is an 82% lifetime risk of breast

cancer (87).

[4] Despite recommendation by the National Institutes of Health Consensus

Conference on Ovarian Cancer, the value of screening with transvaginal

ultrasonography, CA-125 levels, or other procedures is not established in

women at high risk (88). Bourne and coworkers showed that, using this

approach, tumors can be detected approximately 10 times more often than in the

general population, and they recommend screening in high-risk women, but other

groups have not confirmed these findings, and bilateral salpingo-oophorectomy

remains the most effective way to reduce the risk (89).

Data derived from a multicenter consortium of genetic screening centers

indicate that the use of the oral contraceptive pill is associated with a lower

risk for development of ovarian cancer in women who have a mutation in

either BRCA1 or BRCA2 (90,91). The risk reduction is significant: in women

who take oral contraceptives for 5 or more years, the relative risk of ovarian

cancer is 0.4, or a 60% reduction in the incidence of the disease (92).

Prophylactic Salpingo-oophorectomy in High-Risk Women

[6] The value of prophylactic salpingo-oophorectomy in these patients is well

established (93–99). Women at high risk for ovarian cancer who undergo

prophylactic salpingo-oophorectomy have a risk of harboring occult neoplasia: in

one series of 98 such operations, 3 (3.1%) patients were found to have an ovarian

cancer or early invasive cancer in a fallopian tube (96). [6] The protection

against ovarian cancer is high: the performance of a prophylactic salpingooophorectomy reduced the risk of BRCA-related gynecologic cancer by 96%

(96). In a series of 42 such operations, 4 patients (9.5%) had a malignancy, 1 of

which was noted at surgery and 3 that were microscopic; all were smaller than 5

mm (94). Although the risk of ovarian cancer is significantly diminished, there

remains the small risk of peritoneal carcinoma. In these series, the subsequent

development of peritoneal carcinoma was reported to be 0.8% and 1%,

respectively (78,79). The risk of developing subsequent breast cancer may be

reduced by 50% to 80% in BRCA carriers. However, this has been

challenged by a prospective study. In an analysis stratified by BRCA mutation

status and age at diagnosis, they found that oophorectomy was associated with a

significantly reduced risk of breast cancer in BRCA2 carriers diagnosed at age

2567<50 years (age-adjusted hazard ratio [HR], 0.18; p = 0.007) but not among

BRCA1 carriers diagnosed at <50 years (age-adjusted HR, 0.79; p = 0.51). These

findings warrant further evaluation (100).

The role of hysterectomy is more controversial. Most studies show no

increase in the rate of uterine and cervical tumors, but there are rare reports of an

increase of serous tumors of the endometrium. A large multicenter prospective

cohort study concluded that the overall risk of uterine cancer after a risk-reducing

bilateral salpingo-oophorectomy was not increased; however, the risk of serous

and serous-like uterine cancer was increased in BRCA1-positive women, although

still very low, and the benefit of a hysterectomy remains uncertain (101). Women

on tamoxifen are at higher risk for benign endometrial lesions (e.g., polyps) and

endometrial cancer. It is reasonable to consider the performance of a

prophylactic hysterectomy in conjunction with salpingo-oophorectomy in

women with BRCA mutations who are potentially at increased risk of an

endometrial cancer, but this decision should be individualized. The progressionfree survival (PFS) of women who have a BRCA1 or BRCA2 mutation and

develop ovarian cancer is longer than that for those who do not have a mutation.

In one study, the median PFS for mutation carriers was 60 months compared with

22 months for controls. There was no difference in the overall survival between

BRCA mutation carriers and controls (102). BRCA patients have increased

sensitivity to platinum drugs and to the newer agents, poly(ADP-ribose)

polymerase (PARP) inhibitors. [10] The advent of PARP inhibitors and their

increased use for recurrent disease and maintenance therapy, particularly in

BRCA mutation carriers may translate to an improvement in the PFS and

overall survival for BRCA patients.

Recommendations

Recommendations for the management of women at high risk for ovarian cancer

are based on the ACOG practice bulletin no. 182 and summarized as follows:

1. Genetic counseling is recommended for all women with ovarian/fallopian

tube/peritoneal epithelial cancer and for individuals who have a personal

or family history of breast cancer or ovarian cancer. Genetic testing is

recommended when the results of a detailed risks assessment that is performed

as part of genetic counseling suggest the presence of an inherited cancer

syndrome and when the results of testing are likely to influence medical

management.

2. Women who wish to preserve their reproductive capacity can undergo

screening by transvaginal ultrasonography every 6 months or CA-125

measurement and this may be a reasonable short-term surveillance

2568strategy for women at high risk of ovarian cancer starting at age 30 to 35

years until they decide to pursue definitive risk-reducing bilateral

salpingo-oophorectomy.

3. Oral contraceptives should be recommended to young women before they

embark on an attempt to have a family.

4. Women who do not wish to maintain their fertility or who have completed

their families should be recommended to undergo prophylactic bilateral

salpingo-oophorectomy after the age of 35, but by age 40 years. The risk of

ovarian cancers under the age of 40 is very low but the decision regarding the

age of surgery should be based on the age of onset of ovarian cancers in the

family. Most BRCA2-related ovarian cancers tend to occur after the age of 50,

whereas BRCA1-related cancers occur at an earlier age. Typically, riskreducing salpingo-oophorectomy is recommended at age 35 to 40 years

for BRCA1 carriers with the highest lifetime risk of ovarian cancer,

whereas women with BRCA2 may consider delaying until age 40 to 45

years because of later onset of ovarian cancer. These women should be

counseled that this operation does not offer absolute protection, because

peritoneal carcinomas can occur after bilateral salpingo-oophorectomy

(26,29,87).

5. Women with BRCA mutations or who carry another actionable

deleterious mutation that is predisposing to breast cancer should be

offered risk-reducing bilateral mastectomy. Women with BRCA mutations

or who carry another actionable deleterious mutation that is predisposing to

ovarian cancer should be offered risk-reducing bilateral salpingooophorectomy.

6. For a risk-reducing bilateral salpingo-oophorectomy, all tissue from the

ovaries and fallopian tubes should be removed. Thorough visualization of

the peritoneal surfaces should be done and pelvic washings taken. Complete

serial sectioning of the ovaries and fallopian tubes is necessary with

microscopic examination for occult cancer.

7. For women aged 25 to 29 years with known BRCA mutations,

recommended breast cancer surveillance includes clinical breast

examination every 6 to 12 months and annual radiographic evaluation

(preferably magnetic resonance imaging [MRI], with contrast). For

women aged 30 years and older with known BRCA mutations or other

actionable breast cancer mutations, recommended breast cancer

surveillance includes annual mammography and annual breast MRI with

contrast, often alternating every 6 months.

8. Women with a personal or family history of breast or ovarian cancer but

who do not have a documented mutation in BRCA1 or BRCA2 or other

2569hereditary breast- or ovarian-cancer–associated genes should be managed

based on their family history.

9. Women with a documented Lynch syndrome (HNPCC) should undergo

periodic colonoscopy every 1 to 2 years, starting at age 20 to 25 years or 2

to 5 years before the earliest colon cancer diagnosis in the family,

depending on which is earlier. Endometrial biopsy every 1 to 2 years

starting at age 30 to 35 years, is recommended for women with Lynch

syndrome, prophylactic hysterectomy, and bilateral salpingooophorectomy is a risk-reducing option after the completion of

childbearing. In general, risk-reducing hysterectomy and salpingooophorectomy should be discussed with patients by their early to mid-40s

(103,104).

Symptoms

The majority of women with epithelial ovarian cancer have vague and

nonspecific symptoms (4,105). In the early-stage disease, if the patient is

premenopausal, she may experience irregular menses. If a pelvic mass is

compressing the bladder or rectum, she may report urinary frequency or

constipation (105–107). Occasionally, she may perceive lower abdominal

distention, pressure, or pain, such as dyspareunia. Acute symptoms, such as pain

secondary to rupture or torsion, shortness of breath from a malignant pleural

effusion, or symptoms of a bowel obstruction are possible reasons for

presentation.

In advanced-stage disease, patients have symptoms related to the presence

of ascites, omental metastases, or bowel metastases. The symptoms include

abdominal distention, bloating, constipation, nausea, anorexia, or early satiety.

Premenopausal women may report irregular or heavy menses, whereas vaginal

bleeding may occur in postmenopausal women but this is not a typical

presentation of ovarian cancer unless the ovarian tumors are estrogen producing

(107).

Traditionally, ovarian cancer was considered a “silent killer” that did not

produce symptoms until far advanced. Some patients with ovarian cancers

confined to the ovary are asymptomatic, but the majority will have nonspecific

symptoms that do not necessarily suggest an origin in the ovary (107,108–110). In

one survey of 1,725 women with ovarian cancer, 95% recalled symptoms before

diagnosis, including 89% with stages I and II disease and 97% with stages III and

IV disease (107). Some 70% had abdominal or gastrointestinal symptoms, 58%

pain, 34% urinary symptoms, and 26% pelvic discomfort. At least some of these

symptoms could have reflected pressure on the pelvic viscera from the enlarging

ovary. Goff et al. developed an ovarian cancer symptom index and reported that

2570symptoms associated with ovarian cancer, when present for less than 1 year and

occurring longer than 12 days a month, were pelvic/abdominal pain, urinary

frequency/urgency, increased abdominal size or bloating, and difficulty eating or

feeling full (108). The index had a sensitivity of 56.7% for early ovarian cancer

and 79.5% for advanced-stage disease. This study and others suggest that women

and their obstetrician–gynecologist should maintain an appropriate level of

suspicion for further investigation when potential symptoms and signs of ovarian,

fallopian tube, or peritoneal cancer may be present.

A population-based study from Australia found that there did not appear to be a

significant difference in the duration of symptoms or the nature of symptoms in

patients with early as opposed to advanced-stage ovarian cancer, reinforcing the

concept that they are biologically different entities and arguing against the widely

held misconception that early-stage ovarian cancers are at an early stage because

they were diagnosed earlier than patients with more advanced-stage cancers

(109). For fallopian tube carcinoma, the classic triad of symptoms that have been

reported include clear or blood-tinged vaginal discharge, pelvic pain, and a

finding of a pelvic mass. The vaginal discharge termed hydrops tubae profluens

historically has been considered pathognomonic of fallopian tube cancer;

however, in retrospect, these signs are uncommon.

Signs

The most important sign of epithelial ovarian cancer is the presence of a

pelvic mass on physical examination. A solid, irregular, fixed pelvic mass is

highly suggestive of an ovarian malignancy. If an upper abdominal mass or

ascites is present, the diagnosis of ovarian cancer is almost certain. Because the

patient usually reports abdominal symptoms, she may not have a pelvic

examination, and a tumor may be missed.

In patients who are at least 1 year past menopause, the ovaries should be

atrophic and not palpable. It was proposed that any palpable pelvic mass in these

patients should be considered potentially malignant, a situation that was referred

to as the postmenopausal palpable ovary syndrome (111). This concept was

challenged, because subsequent authors reported that only about 3% of palpable

masses measuring less than 5 cm in postmenopausal women are malignant (63).

Atypical glandular cells on cervical pathology may signal an upper genital tract

malignancy. This finding on a Papanicolaou (Pap) test should prompt further

evaluation for cervical or endometrial carcinoma. If there are no findings to

suggest these cancers, evaluation should continue for other malignancies of the

female genital tract. In a systematic review of approximately 7,000 women with

atypical glandular cells on a Pap test only 5.2% of patients had a malignancy.

Among patients with a malignancy, there were 5.4% ovarian cancers and 1.0%

2571fallopian tube cancers (112).

Women with ovarian cancer can rarely present with signs of a paraneoplastic

syndrome, which may include disseminated intravascular coagulation,

polyneuritis, dermatomyositis, hemolytic anemia, and cerebellar degeneration

(113).

Diagnosis

Ovarian epithelial cancers must be differentiated from benign neoplasms

and functional cysts of the ovaries. A variety of benign conditions of the

reproductive tract, such as pelvic inflammatory disease, endometriosis, and

pedunculated uterine leiomyomas, can simulate ovarian cancer. Nongynecologic

causes of a pelvic tumor, such as an inflammatory (e.g., diverticular) disease or

neoplastic colonic mass, must be excluded (4). A pelvic kidney can simulate

ovarian cancer.

Serum CA-125 levels are useful in distinguishing malignant from benign

pelvic masses (114). For a postmenopausal patient with an adnexal mass and

a very high serum CA-125 level (>200 U/mL), there is a 96% positive

predictive value for malignancy. For premenopausal patients, the specificity

of the test is low because the CA-125 level tends to be elevated in common

benign conditions.

For the premenopausal patient, a period of observation is reasonable,

provided the adnexal mass does not have characteristics that suggest

malignancy (i.e., it is mobile, mostly cystic, unilateral, and of regular

contour). An interval of no more than 2 months is allowed, during which

hormonal suppression with an oral contraceptive may be used. If the lesion is not

neoplastic, it should regress, as measured by pelvic examination and pelvic

ultrasonography. If the mass does not regress or if it increases in size, it must be

presumed to be neoplastic and must be removed surgically.

The size of the lesion is important. If a cystic mass is greater than 8 cm in

diameter, the probability is high that the lesion is neoplastic, unless the patient is

taking clomiphene citrate or other agents to induce ovulation (115–118).

Premenopausal patients whose lesions are clinically suspicious (i.e., large,

predominantly solid, relatively fixed, or irregularly shaped) should undergo

laparoscopy or laparotomy with keen attention to avoid intraoperative

rupture or spillage, as should postmenopausal patients with complex adnexal

masses of any size. Image-guided biopsy is not performed for fear of rupture

and seeding a potential malignancy.

Ultrasonographic signs of malignancy include an adnexal pelvic mass with

areas of complexity, such as irregular borders, multiple echogenic patterns

within the mass, and dense multiple irregular septae. Bilateral tumors are

2572more likely to be malignant, although the individual characteristics of the lesions

are of greater significance. Transvaginal ultrasonography may have a somewhat

better resolution than transabdominal ultrasonography for adnexal neoplasms

(119–122). Doppler color flow imaging may enhance the specificity of

ultrasonography for demonstrating findings consistent with malignancy (123–

125).

In postmenopausal women with unilocular cysts measuring 8 to 10 cm or

less and normal serial CA-125 levels, expectant management is acceptable,

and this approach may decrease the number of surgical interventions (126–128).

The suspicion of ovarian cancer or the diagnosis of an ovarian cancer

requires surgical exploration by a gynecologic oncologist, which may be by

an exploratory laparotomy or a minimally invasive technique such as

laparoscopy or a robotic platform. The preoperative evaluation of the patient

with an adnexal mass is outlined in Figure 10.2 (see Chapter 10, page 201).

Before the planned exploration, the patient should undergo routine hematologic

and biochemical assessments. A preoperative evaluation in a patient undergoing

laparotomy should include a radiograph of the chest. Abdominal and pelvic

computed tomography (CT) or magnetic resonance imaging (MRI) are of limited

value for a patient with a definite pelvic mass (129–131). A CT or MRI should be

performed for patients with ascites and no pelvic mass to look for liver or

pancreatic tumors. The findings only rarely preclude laparotomy (129). The value

of PET scanning is still being evaluated (131–133). If the hepatic enzyme values

are normal, the likelihood of liver disease is low. Liver–spleen scans, bone scans,

and brain scans are unnecessary unless symptoms or signs suggest metastases to

these sites.

The preoperative evaluation should exclude other primary cancers

metastatic to the ovary. A barium enema or colonoscopy is indicated in selected

patients with symptoms and signs suspicious for colon cancer. This study should

be performed for any patient who has evidence of occult blood in the stool or has

signs of lower intestinal obstruction. An upper gastrointestinal radiographic series

or gastroscopy is indicated if there are upper gastrointestinal symptoms such as

nausea, vomiting, or hematemesis (4,134). Bilateral diagnostic mammography is

indicated if there is any breast mass, because breast cancer metastatic to the

ovaries can simulate primary ovarian cancer.

A Pap test can be performed, although its value for the detection of ovarian

cancer is very limited. Patients who have irregular menses or postmenopausal

vaginal bleeding should have endometrial biopsy and endocervical curettage to

exclude the presence of uterine or endocervical cancer metastatic to the ovary.

Differential Diagnosis

2573Ovarian epithelial cancers must be differentiated from benign neoplasms

and functional cysts of the ovaries (126–128). A variety of benign conditions

of the reproductive tract, such as pelvic inflammatory disease, endometriosis,

and pedunculated uterine leiomyomata, can simulate ovarian cancer.

Nongynecologic causes of a pelvic tumor, such as an inflammatory or neoplastic

colonic mass, must be excluded. Gastrointestinal and breast cancers are the most

common nongenital malignancies that metastasize to the ovary (135,136).

Patterns of Spread

Ovarian epithelial cancers spread primarily by exfoliation of cells into the

peritoneal cavity, lymphatic dissemination, and hematogenous spread.

Transcoelomic

The most common and earliest mode of dissemination of ovarian epithelial

cancer is by exfoliation of cells that implant along the surfaces of the

peritoneal cavity. The cells tend to follow the circulatory path of the peritoneal

fluid. The fluid moves with the forces of respiration from the pelvis, up the

paracolic gutters, especially on the right, along the intestinal mesenteries, to the

right hemidiaphragm. Metastases are typically seen on the posterior cul-de-sac,

paracolic gutters, right hemidiaphragm, liver capsule, the peritoneal surfaces of

the intestines and their mesenteries, and the omentum. The disease seldom

invades the intestinal lumen but progressively agglutinates loops of bowel,

leading to a functional intestinal obstruction. This condition is known as

carcinomatous ileus (4).

Lymphatic

Lymphatic dissemination to the pelvic and para-aortic lymph nodes is

common, particularly in advanced-stage disease (137–139). Spread through the

lymphatic channels of the diaphragm and through the retroperitoneal lymph nodes

can lead to dissemination above the diaphragm, especially to the supraclavicular

lymph nodes (137). Burghardt et al. reported that 78% of patients with stage III

disease have metastases to the pelvic lymph nodes (139). In another series, the

rate of para-aortic lymph nodes positive for metastasis was 18% in stage I, 20% in

stage II, 42% in stage III, and 67% in stage IV (137).

Hematogenous

Hematogenous dissemination at the time of diagnosis is uncommon. Spread to

vital organ parenchyma, such as the lungs and liver, occurs in only about 2% to

3% of patients. Most patients with disease above the diaphragm when diagnosed

2574have a right pleural effusion (4). Systemic metastases appear more frequently in

patients who survived for some years. Dauplat et al. reported that distant

metastasis consistent with stage IV disease ultimately occurred in 38% of the

patients whose disease was originally IP (140).

Prognostic Factors

The outcome of treatment can be evaluated in the context of prognostic

factors, which can be grouped into pathologic, biologic, and clinical factors

(141).

Pathologic Factors

The morphology and histologic pattern, including the architecture and grade

of the lesion, are important prognostic variables (4). The histologic type was

not believed to have prognostic significance, but several papers contained

suggestions that clear cell carcinomas are associated with a prognosis worse than

that of other histologic types (141,142).

Histologic grade, as determined either by the pattern of differentiation or by the

extent of cellular anaplasia and the proportion of undifferentiated cells, seems to

be of prognostic significance (143–146). Studies of the reproducibility of grading

ovarian cancers show a high degree of intraobserver and interobserver variation

(147,148). Baak et al. have presented a standard grading system based on the

morphometric analysis, and the system seems to correlate with prognosis,

especially in its ability to distinguish low-grade or borderline patterns from other

tumors (149).

Clinical Factors

In addition to the stage, the extent of residual disease after primary surgery,

the volume of ascites, patient age, and performance status are all

independent prognostic variables (150–159). Among patients with stage I

disease, Dembo et al. showed, in a multivariate analysis, that tumor grade and

dense adherence to the pelvic peritoneum had a significant adverse impact on

prognosis, whereas intraoperative tumor spillage or rupture did not (156). Sjövall

et al. confirmed that ovarian cancers that undergo intraoperative rupture or

spillage do not worsen prognosis, whereas tumors that are ruptured

preoperatively do have a poorer prognosis (157). A multivariate analysis of

these and several other studies was performed by Vergote et al., who found that

for early-stage disease, poor prognostic variables were tumor grade, capsular

penetration, surface excrescences, and malignant ascites, but not iatrogenic

rupture (159).

2575Table 39-2 2013 FIGO Staging for Primary Carcinoma of the Ovary, Fallopian Tube,

or Primary Peritoneal Cancer

Stage I Growth Limited to the Ovaries or Fallopian Tubes

IA Growth limited to one ovary or fallopian; no ascites containing

malignant cells

No tumor on the external surface; capsule intact

IB Growth limited to both ovaries or fallopian tubes; no ascites containing

malignant cells

No tumor on the external surfaces; capsules intact

IC Tumor limited to one or both ovaries or fallopian tubes with any of the

following:

IC1 Surgical spill

IC2 Capsule ruptured before surgery or tumor on ovarian or fallopian tube

surface

IC3 Malignant cells in the ascites or peritoneal washings

Stage II Growth involving one or both ovaries or fallopian tubes with pelvic

extension (below the pelvic brim)

IIA Extension and/or metastases to the uterus and/or fallopian tubes and/or

ovaries

IIB Extension to other pelvic tissues

Stage III Tumor involving one or both ovaries or fallopian tubes or

peritoneal cancer with cytologically or histologically confirmed

spread to the peritoneum outside the pelvis and/or metastasis to the

retroperitoneal lymph nodes

IIIA Positive retroperitoneal lymph nodes and/or microscopic metastasis

beyond pelvis

IIIA1(i) Positive retroperitoneal lymph nodes with metastasis up to 10 mm in

greatest dimension

IIIA1(ii) Positive retroperitoneal lymph nodes with metastasis more than 10 mm

in greatest dimension

2576IIIA2 Microscopic extrapelvic (above the pelvic brim) peritoneal

involvement, with or without positive retroperitoneal lymph nodes

IIIB Macroscopic peritoneal metastasis beyond pelvis up to 2 cm in greatest

dimension, with or without positive retroperitoneal nodes

IIIC Macroscopic peritoneal metastasis beyond pelvis more than 2 cm in

greatest dimension (includes extension of tumor to capsule of liver and

spleen without parenchymal involvement of either organ), with or

without positive retroperitoneal nodes

IV Distant metastasis (excludes peritoneal metastasis)

IVA Pleural effusion is present, there must be positive cytologic test

results

IVB Parenchymal metastases and metastases to extra-abdominal organs

(including lymph nodes and lymph nodes outside the abdominal

cavity)

The histologic characteristics are to be considered in the staging, as are results of cytologic

testing as far as effusions are concerned. It is desirable that a biopsy be performed on

suspicious areas outside the pelvis.

FIGO, International Federation of Obstetrics and Gynecology.

aDense adhesions with histologically proven tumor cells justify upgrading to stage II.

Transmural bowel infiltration or umbilical deposit are stage IVB.

Initial Surgery for Ovarian Cancer

Staging

Ovarian epithelial malignancies are staged according to the FIGO system

listed in Table 39-2 (160). The FIGO staging is based on findings at surgical

exploration. A preoperative evaluation should exclude the presence of

extraperitoneal metastases.

[7] Thorough surgical staging and search for microscopic metastasis should

be performed when there is no visible or palpable disease in the intraabdominal space outside the ovaries and fallopian tubes. Subsequent

treatment may be determined by the stage of the disease. In earlier series in

which patients did not undergo careful surgical staging, the overall 5-year

survival for patients with apparent stage I epithelial ovarian cancer was only

2577about 60% (161). Survival rates of 90% to 100% are reported for patients who

were properly staged and were found to have stage IA or IB disease.

Technique for Surgical Staging

In patients whose preoperative evaluation suggests a probable malignancy, a

vertical abdominal incision is recommended to allow adequate access to the

upper abdomen (4,161). When a malignancy is unexpectedly discovered in a

patient who has a lower transverse incision, the rectus muscles can be either

divided or detached from the symphysis pubis to allow better access to the upper

abdomen. If this is not sufficient, the incision can be extended on one side to

create a “J” incision (4).

The ovarian tumor should be removed intact, if possible, and a frozen

histologic section should be obtained. A thorough surgical staging should be

performed. Staging involves the following steps (4,161):

1. Any free fluid, especially in the pelvic cul-de-sac, should be submitted for

cytologic evaluation.

2. If no free fluid is present, peritoneal washings should be performed by

instilling and recovering 50 to 100 mL of saline from the pelvic cul-de-sac,

each paracolic gutter, and beneath each hemidiaphragm. Obtaining the

specimens from under the diaphragms can be facilitated with the use of a

rubber catheter attached to the end of a bulb syringe.

3. A systematic exploration of all the intra-abdominal surfaces and viscera is

performed, proceeding in a clockwise fashion from the cecum cephalad along

the paracolic gutter and the ascending colon to the right kidney, the liver and

gallbladder, the right hemidiaphragm, the entrance to the lesser sac at the paraaortic area, across the transverse colon to the left hemidiaphragm, down the

left gutter and the descending colon to the rectosigmoid colon. The small

intestine and its mesentery from the Treitz ligament to the cecum should be

inspected.

4. Any suspicious areas or adhesions on the peritoneal surfaces should be

biopsied. If there is no evidence of disease, multiple IP biopsies should be

performed. Tissue from the peritoneum of the pelvic cul-de-sac, both paracolic

gutters, the peritoneum over the bladder, and the intestinal mesenteries should

be taken for biopsy.

5. The diaphragm should be sampled, either by biopsy or by scraping with a

tongue depressor, and a sample obtained for cytologic assessment.

Biopsies of any irregularities on the surface of the diaphragm can be facilitated

by the use of the laparoscope and the associated biopsy instrument.

6. The omentum should be resected from the transverse colon, including

2578from the hepatic and splenic flexure of the colon and separated from its

attachment to the greater curvature of the stomach, including ligation of

the left and right gastroepiploic and short gastric arteries, a procedure

called supracolic omentectomy. The procedure is initiated on the underside of

the greater omentum, where the peritoneum is incised a few millimeters away

from the transverse colon. The branches of the gastroepiploic vessels are

clamped, ligated, and divided, along with all the small branching vessels that

feed the infracolic omentum.

7. The retroperitoneal spaces should be explored to evaluate the pelvic and

para-aortic lymph nodes. The retroperitoneal dissection is performed by

incision of the peritoneum over the psoas muscles. Any enlarged lymph nodes

should be resected and submitted for frozen section. If no metastases are

present, a formal pelvic lymphadenectomy should be performed. The paraaortic area should be palpated and lymph nodes sampled if enlarged or

suspicious.

Results

Metastases in apparent stages I and II epithelial ovarian cancer occur in as

many as 3 in 10 patients whose tumors appear to be confined to the pelvis

but who have occult metastatic disease in the upper abdomen or the

retroperitoneal lymph nodes (138,162–169). In a literature review, occult

metastases in such patients were found in biopsies of the diaphragm in 7.3%,

biopsies of the omentum in 8.6%, the pelvic lymph nodes in 5.9%, the aortic

lymph nodes in 18.1%, and in 26.4% of peritoneal washings (161).

The importance of careful initial surgical staging is emphasized by the findings

of a cooperative national study in which 100 patients with apparent stages I and II

disease were referred for subsequent therapy and underwent additional surgical

staging (162). In this series, 28% of the patients initially believed to have stage I

disease were upstaged and 43% of those believed to have stage II disease had

more advanced lesions. A total of 31% of the patients were upstaged as a

result of additional surgery, and 77% were reclassified as having actual stage

III disease. Histologic grade was a significant predictor of occult metastasis.

Sixteen percent of the patients with grade 1 lesions were upstaged, compared with

34% with grade 2 disease and 46% with grade 3 disease.

Borderline Tumors

The principal treatment of borderline (low malignant potential) ovarian

tumors is surgical resection of the primary tumor. There is no evidence that

either subsequent chemotherapy or radiation therapy improves survival. When a

frozen section determines that the histology is borderline, premenopausal

2579patients who desire preservation of the ovarian function may undergo a

conservative operation, a unilateral oophorectomy (4,170). In a study of

patients who underwent unilateral ovarian cystectomy for apparent stage I

borderline serous tumors, Lim-Tan et al. found that this conservative operation

was safe, with only 8% of the patients developing recurrences 2 to 18 years later,

all with curable disease confined to the ovaries (170). Recurrence was associated

with positive margins of the removed ovarian cyst. Thus, hormonal function and

fertility can be maintained (4,170). For patients who had an oophorectomy or

cystectomy and a borderline tumor is documented later in the permanent

pathology, no additional immediate surgery is necessary.

Stage I

After a comprehensive staging laparotomy, only a minority of women will

have local disease (FIGO stage I). There are over 20,000 women diagnosed

yearly with epithelial ovarian cancer in the United States, and nearly 4,000 of

these have disease confined to the ovaries (65,171). The prognosis for these

patients depends on the clinical–pathologic features. Because of this emphasis on

the importance of surgical staging, the rate of lymph node sampling increased in

the United States, with a study showing that for women with stages I and II

disease, the percentage having lymph nodes sampled increased from 38% to 59%

from 1991 to 1996 (172).

The primary surgical treatment for stage I epithelial ovarian cancer is

surgical, and patients should undergo total abdominal hysterectomy,

bilateral salpingo-oophorectomy, and surgical staging (161,162). In certain

circumstances, a unilateral salpingo-oophorectomy may be performed as a

part of fertility-sparing surgery. Based on the findings at surgery and the

pathologic evaluation, patients with stage I ovarian cancer can be grouped into

low-risk and high-risk categories (Table 39-3).

Table 39-3 Prognostic Variables in Early-Stage Epithelial Ovarian Cancer

Low Risk High Risk

Low grade High grade

Intact capsule Tumor growth through capsule

No surface excrescences Surface excrescences

No ascites Ascites

Negative peritoneal cytologic findings Malignant cells in fluid

2580Unruptured or intraoperative rupture Preoperative rupture

No dense adherence Dense adherence

Diploid tumor Aneuploid tumor

From Berek JS, Friedlander ML, Hacker NF. Epithelial ovarian, fallopian tube, and

peritoneal cancer. In: Berek JS, Hacker NF. Berek & Hacker’s Gynecologic Oncology. 6th

ed. Philadelphia, PA: Wolters Kluwer; 2015:483.

Stage I Low Risk

Fertility Preservation in Early-Stage Ovarian Cancer

For patients who underwent a thorough staging laparotomy and who have

no evidence of spread beyond the ovary, abdominal hysterectomy and

bilateral salpingo-oophorectomy are appropriate therapy. The uterus and

the contralateral ovary can be preserved in women with stage IA, grade 1 to

2 disease who desire to preserve fertility. The women undergoing disease

surveillance should be monitored carefully with routine periodic pelvic

examinations and determinations of serum CA-125 levels. The other ovary and

the uterus are removed at the completion of childbearing.

Guthrie et al. studied the outcome of 656 patients with early-stage epithelial

ovarian cancer (169). No untreated patients who had stage IA, grade 1 cancer died

of their disease; thus, adjuvant radiation and chemotherapy are unnecessary. The

Gynecologic Oncology Group (GOG) carried out a prospective, randomized trial

of observation versus melphalan for patients with stages IA and IB, grade 1 or 2

disease (142). Five-year survival for each group was 94% and 96%, respectively,

confirming that no further treatment is needed for such patients.

Stage I High Risk

Patients who have more poorly differentiated disease or who have malignant

cells either in ascites fluid or in peritoneal washings, must undergo complete

surgical staging (4). Most patients require chemotherapy.

Advanced-Stage Ovarian Cancer

The management of all patients with advanced-stage disease is approached in

a similar manner, with modifications made in response to the overall status

and general health of the patient and the extent of residual disease present at

the time treatment is initiated. A treatment scheme is outlined in Figure 39-

10. Most patients subsequently receive combination chemotherapy for an

empiric number of cycles.

2581Cytoreductive Surgery for Advanced-Stage Disease

If the patient is medically stable, she should undergo cytoreductive surgery

to remove as much of the tumor and its metastases as possible (173–200). The

operation to remove the primary tumor and the associated metastatic disease

is referred to as debulking or cytoreductive surgery. The operation typically

includes the performance of a total abdominal hysterectomy and bilateral

salpingo-oophorectomy, along with a complete omentectomy and resection of any

metastatic lesions from the peritoneal surfaces or from the intestines. The pelvic

tumor often directly involves the rectosigmoid colon, the terminal ileum, and the

cecum (Fig. 39-11). In a minority of patients, most or all of the disease is

confined to the pelvic viscera and the omentum, so that removal of these organs

will result in extirpation of all gross tumor, a situation that is associated with a

reasonable chance of prolonged PFS.

The paradigm of routine lymphadenectomy in patients with advanced ovarian

cancer and clinically and radiographically negative nodes has been challenged

with the reporting of the Lymphadenectomy in Ovarian Neoplasms (LION) study.

This was a prospective randomized trial of 647 patients with stage IIB to IV

ovarian cancer with macroscopic complete resection and preoperative and

intraoperative clinically negative lymph nodes randomized intraoperatively to

systematic pelvic and para-aortic lymphadenectomy versus no lymphadenectomy.

This trial showed that systematic pelvic and para-aortic lymphadenectomy in

patients with advanced ovarian cancer with intra-abdominal complete resection

and clinically negative lymph nodes does not provide a PFS or overall survival

benefit, despite the detection and removal of subclinical lymph nodal metastases

in approximately 56% of patients. The authors suggest that routine

lymphadenectomy should be omitted in these patients to reduce postoperative

morbidity and mortality (201).

The removal of bulky tumor masses may reduce the volume of ascites

present. Often, ascites will disappear after removal of the primary tumor and a

large omental “cake.” Removal of the omental cake may alleviate the nausea and

early satiety that many patients experience. Removal of intestinal metastases may

restore adequate intestinal function and improve the overall nutritional status of

the patient, thereby facilitating the patient’s ability to tolerate subsequent

chemotherapy.

A large, bulky tumor may contain areas that are poorly vascularized, and

these areas will be exposed to suboptimal concentrations of

chemotherapeutic agents. Similarly, these areas are poorly oxygenated, so that

radiation therapy, which requires adequate oxygenation to achieve maximal cell

kill, will be less effective. Surgical removal of these bulky tumors may eliminate

areas that could be relatively resistant to radiation and chemotherapeutic

2582treatment.

Larger tumor masses tend to be composed of a higher proportion of cells that

are either nondividing or in the “resting” phase (i.e., G0 cells, which are

essentially resistant to the therapy). A low growth fraction is characteristic of

bulky tumor masses, and cytoreductive surgery can result in smaller residual

masses with a relatively higher growth fraction.

Goals of Cytoreductive Surgery

The principal goal of cytoreductive surgery is removal of all of the primary

cancer and, if possible, all metastatic disease. If resection of all metastases is

not feasible, the goal is to reduce the tumor burden by resection of all individual

tumors to an optimal status (<1 cm of any single implant of residual disease).

Patients whose disease is completely resected to no macroscopic (microscopic

only) residual disease have the best overall survival (Fig. 39-12) (178).

Approximately 30% to 40% of patients in this category will be free of disease

at 5 years.


FIGURE 39-10 Treatment scheme for patients with advanced-stage ovarian cancer. *In

selected cases of stage IIIC/IV disease, neoadjuvant chemotherapy may be given, and then

an interval cytoreductive surgery is performed after three cycles. +Chemotherapy depends

on whether platinum-sensitive or platinum-resistant. (Modified from Berek JS,

Friedlander ML, Hacker NF. Epithelial ovarian, fallopian tube, and peritoneal cancer. In:

Berek JS, Hacker NF. Berek & Hacker’s Gynecologic Oncology. 6th ed. Philadelphia, PA:

Wolters Kluwer; 2015:485.)

The resectability of the metastatic tumor is usually determined by the

location of the disease. Optimal cytoreduction is difficult to achieve in the

presence of extensive disease on the diaphragm, in the parenchyma of the liver,

along the base of the small bowel mesentery, in the lesser omentum, or in the

porta hepatis. [7] Neoadjuvant chemotherapy is an option for patients with

advanced ovarian/fallopian tube or peritoneal cancer who are determined to

have a low likelihood of optimal cytoreduction or in women determined to

have a high perioperative risk (202).

In two published large randomized control trials, neoadjuvant chemotherapy

was noninferior to upfront cytoreduction (203,204). There was no significant

difference between the PFS or overall survival between the two groups. In one

trial, the EORTC 55971, outcomes were found to vary by the stage of disease and

tumor size. In particular, patients with stage IV disease and more extensive

tumors greater than 45 mm had a better survival with neoadjuvant chemotherapy;

however, patients with stage IIIC cancer and metastatic lesions less than 45 mm

had a better survival with primary cytoreductive surgery (205). All randomized

clinical trials have shown lower perioperative morbidity with neoadjuvant

chemotherapy compared to primary cytoreductive surgery (202).


FIGURE 39-11 Extensive ovarian carcinoma involving the bladder, rectosigmoid, and

ileocecal area. (Redrawn from Heintz APM, Berek JS. Cytoreductive surgery for ovarian

carcinoma. In: Piver MS, ed. Ovarian Malignancies. Edinburgh, UK: Churchill

Livingstone; 1987:134, with permission.)


FIGURE 39-12 Survival of patients with stage IIIC epithelial ovarian cancer based on the

maximal size of residual tumor after exploratory laparotomy and tumor resection. (From

Heintz APM, Odicino F, Maisonneuve P, et al. Carcinoma of the ovary. Twenty-sixth

annual report of the results of treatment of gynaecological cancer. Int J Gynecol Oncol

2006;95(suppl 1): S161–S192, with permission.)

Exploration

The supine position on the operating table may be sufficient for surgical

exploration of most patients. For patients with extensive pelvic disease and for

whom a low resection of the colon may be necessary, the low lithotomy position

should be used. Debulking operations should be performed through a vertical

incision to gain adequate access to the upper abdomen and to the pelvis.

After the peritoneal cavity is opened, ascites fluid, if present, should be

evacuated. In some centers, fluid is submitted for in vitro research studies, such

as molecular analyses. In cases of massive ascites, careful attention must be given

to hemodynamic monitoring, especially for patients with borderline

cardiovascular function.

The peritoneal cavity and retroperitoneum are thoroughly inspected and

palpated to assess the extent of the primary tumor and the metastatic

2587disease. All abdominal viscera must be palpated to exclude the possibility that the

ovarian disease is metastatic, particularly from the stomach, colon, or pancreas. If

optimal cytoreduction is not considered achievable, extensive bowel and urologic

resections are not indicated, except to overcome a bowel obstruction. Removal of

the primary tumor and omental cake is usually feasible and desirable.

Pelvic Tumor Resection

The essential principle of pelvic tumor removal is the retroperitoneal

approach. To accomplish this, the retroperitoneum is entered laterally, along the

surface of the psoas muscles, which avoids the iliac vessels and the ureters. If the

uterus is present, the procedure is initiated by bilateral division of the round

ligaments. The peritoneal incision is extended cephalad, lateral to the ovarian

vessels within the infundibulopelvic ligament, and caudally toward the bladder.

With careful dissection, the retroperitoneal space is explored, and the ureter and

pelvic vessels are identified. The pararectal and paravesical spaces are identified

and developed, as described in Chapter 38.

The peritoneum overlying the bladder is dissected to connect the peritoneal

incisions anteriorly. The vesicouterine plane is identified, and with careful sharp

dissection the bladder is mobilized from the anterior surface of the cervix. The

ovarian vessels are isolated, doubly ligated, and divided.

Hysterectomy is performed. The ureters must be carefully displayed to avoid

injury. During this procedure, the uterine vessels can be identified. Ligation of the

uterine vessels and the remainder of the tissues within the cardinal ligaments

completes the hysterectomy and resection of the contiguous tumor.

Because epithelial ovarian cancers tend not to invade the lumina of the

colon or bladder, it is usually feasible to resect pelvic tumors without having

to resect portions of the lower colon or the urinary tract (183,184). Resection

of a small portion of the bladder may be required and, if so, a cystotomy should

be performed to assist in resection of the disease (184).

Intestinal Resection

Resection of focal areas of disease involving the small or large intestine

should be performed if that would permit the removal of all or most of the

abdominal metastases and leave the patient with optimal cytoreduction at the

end of surgery. Apart from the rectosigmoid colon, the most frequent sites of

intestinal metastasis are the terminal ileum, the cecum, and the transverse colon.

Resection of one or more of these segments of bowel may be necessary

(183,185).

If the disease surrounds the rectosigmoid colon and its mesentery, that portion

of the colon may have to be removed in order to clear the pelvic disease (Fig. 39-

258813) (183). After the pararectal space is identified, the proximal site of colonic

involvement is identified, the colon and its mesentery are divided, and the

rectosigmoid is removed along with the uterus en bloc. A reanastomosis of the

colon is performed.

Omentectomy

Advanced epithelial ovarian cancer often completely replaces the omentum,

forming an “omental cake.” This disease may be adherent to the parietal

peritoneum of the anterior abdominal wall, making entry into the abdominal

cavity difficult. After freeing the omentum from any adhesions to the parietal

peritoneum, adherent loops of the small intestine are freed by sharp dissection.

The omentum is lifted and pulled gently in the cranial direction, exposing the

attachment of the infracolic omentum to the transverse colon. The peritoneum is

incised to open the appropriate plane, which is developed by sharp dissection

along the serosa of the transverse colon. Small vessels are ligated with hemoclips,

suture, or a vessel-sealing device. The omentum is separated from the greater

curvature of the stomach by ligation of the right and left gastroepiploic arteries

and ligation of the short gastric arteries (Fig. 39-14).

The disease in the gastrocolic ligament can extend to the hilus of the spleen

and splenic flexure of the colon on the left and to the capsule of the liver and

the hepatic flexure of the colon on the right. Usually, the disease does not

invade the parenchyma of the liver or spleen, and a plane can be found between

the tumor and these organs. It will occasionally be necessary to perform

splenectomy to remove all the omental disease (186).

Resection of Other Metastases

Other large masses of tumor located on the parietal peritoneum should be

removed, particularly if they are isolated masses and their removal will

permit optimal cytoreduction. Resection of extensive disease from the surfaces

of the diaphragm is neither practical nor feasible, although solitary metastases

may be resected, the diaphragm sutured, and a chest tube placed for a few days if

needed (186,187). The use of the Cavitron Ultrasonic Surgical Aspirator (CUSA)

and the argon beam coagulator may facilitate resection of small tumor nodules,

especially those on flat surfaces (188,189).

Feasibility and Outcome

There was no randomized prospective study performed to define the value of

primary cytoreductive surgery, but all retrospective studies indicate that the

diameter of the largest residual tumor nodule before the initiation of

chemotherapy is significantly related to the PFS in patients with advanced ovarian

2589cancer (190). The quality of life may be significantly enhanced by the removal of

bulky tumor masses from the pelvis and upper abdomen (191).

An analysis of the retrospective data indicates that, when performed by

gynecologic oncologists, successful operations are feasible in 70% to 90% of

patients (181,182). Major morbidity is approximately 5% and operative mortality

is 1% (185,192,193). Intestinal resection in these patients does not appear to

increase the overall morbidity caused by the operation (185).


FIGURE 39-13 The resection of the pelvic tumor may include removal of the uterus,

2590tubes, and ovaries, as well as portions of the lower intestinal tract. The arrows represent

the plane of resection. (Modified from Berek JS, Friedlander ML, Hacker NF. Epithelial

ovarian, fallopian tube, and peritoneal cancer. In: Berek JS, Hacker NF. Berek & Hacker’s

Gynecologic Oncology. 6th ed. Philadelphia, PA: Wolters Kluwer; 2015:490.)


FIGURE 39-14 Separation of the omentum from stomach and transverse colon. Arrow, the

direction of the initial surgical approach. (From Heintz APM, Berek JS. Cytoreductive

surgery for ovarian carcinoma. In: Piver MS, ed. Ovarian Malignancies. Edinburgh, UK:

Churchill Livingstone; 1987:134, with permission.)

In a meta-analysis of 81 studies of women who underwent cytoreductive

surgery for advanced ovarian cancer, Bristow et al. documented that the

extent of debulking correlated with incremental benefits in survival (i.e., the

greater the percentage of tumor reduction, the longer the survival). Each

10% increase in cytoreduction equaled a 5.5% increase in median survival

(190). Women whose cytoreduction was greater than 75% of their tumor

burden had a median survival of 33.9 months compared with 22.7 months for

women whose tumors were cytoreduced less than 75% (p <0.001). The

performance of a pelvic and para-aortic lymphadenectomy in patients with stage

III disease does not prolong survival, based on the results of a large prospective,

2591randomized trial (194).

A prospective randomized study of “interval” cytoreductive surgery was

carried out by the European Organization for the Research and Treatment

of Cancer (EORTC). Interval surgery was performed after three cycles of

platinum-combination chemotherapy in patients whose primary attempt at

cytoreduction was suboptimal. The initial surgery for most of these patients was

not an aggressive attempt to debulk their tumors. Patients in the surgical arm of

the study demonstrated a survival benefit when compared with those who did not

undergo interval debulking (195). The risk of mortality was reduced by more than

40% in the group that was randomized to the debulking arm of the study. Based

on these data, the performance of a debulking operation as early as possible in the

course of the patient’s treatment should be considered the standard of care (191).

A prospective phase III study of interval cytoreductive surgery was

conducted by the GOG; the patients entered on the trial had a maximal

attempt at tumor resection at their initial surgery (196). The randomized

findings showed no difference between the patients who had an additional attempt

at debulking after three cycles of chemotherapy compared with those who did not.

The median survival of the 216 women who underwent interval cytoreduction

was 32 months compared with 33 months for the 209 women who did not

undergo surgical cytoreduction.

There is evidence that the survival of women with advanced ovarian cancer is

improved when the surgeon is specifically trained to perform cytoreductive

surgery and when there is centralization of care (197–200). Whenever feasible,

patients with advanced ovarian malignancy should be referred to a

subspecialty unit for primary surgery, and every effort should be made to

attain as complete a cytoreduction as possible.

Chemotherapy

Stage I Epithelial Ovarian Cancer

Early Stage, Low Risk

Guthrie et al. studied the outcome of 656 patients with early-stage epithelial

ovarian cancer (169). Patients who had stage IA, grade 1 cancer and did not

receive radiation or chemotherapy did not die of their disease; indicating that

adjuvant therapy is unnecessary. The GOG carried out a prospective, randomized

trial of observation versus melphalan for patients with stages IA and IB, grades 1

and 2 disease (142). Five-year survival for each group was 94% and 96%,

respectively, confirming that adjuvant treatment did not improve survival.

Therefore, no adjuvant chemotherapy is recommended for these patients.

2592Early Stage, High Risk

In patients whose disease is high risk (e.g., high-grade serous or in whom

there are malignant cells either in ascites fluid or in peritoneal washings),

additional therapy is indicated. Chemotherapy is recommended for these

patients to reduce recurrence risk (206–219).

Two large parallel randomized phase III clinical trials were conducted on

women with early-stage disease more than 20 years ago and reported in 2003: the

International Collaborative Ovarian Neoplasm Trial 1 (ICON1) and the

Adjuvant Chemotherapy Trial in Ovarian Neoplasia (ACTION) (220,221).

In the ICON1 trial, 477 patients from 84 centers in Europe were entered.

Patients of all stages were eligible for the trial if, in the opinion of the

investigator, it was unclear whether adjuvant therapy would be of benefit. Most

patients were considered to have stages I and IIA disease, but optimal surgical

staging was not required, and it is likely that a significant number of these

women had stage III disease. Adjuvant platinum-based chemotherapy was given

to 241 patients, and no adjuvant chemotherapy was given to 236 patients. The 5-

year survival was 73% in the group who received adjuvant chemotherapy

compared with 62% in the control group (HR, 0.65; p = 0.01) (221).

In the ACTION trial, 440 patients from 40 European centers were

randomized; 224 patients received adjuvant platinum-based chemotherapy, and

224 patients did not (220). Patients with stages I and II, grades 2 and 3 were

eligible. Only about one-third of the total group was optimally staged (151

patients). In the observation arm, optimal staging was associated with a

better survival (HR, 2.31; p = 0.03), and in the suboptimally staged patients,

adjuvant chemotherapy was associated with an improvement in survival

(HR, 1.78; p = 0.009). In the ACTION trial, the initial trial results suggested

that the benefit from adjuvant chemotherapy was limited to the patients with

suboptimal staging. However, a long-term follow-up of the randomized trial

after a median of 10.1 years showed that the overall survival after optimal

staging was improved even among those who received adjuvant

chemotherapy (HR of death, 1.89; p = 0.05) (222).

When the data from the two trials were combined and analyzed, a total of 465

patients were randomized to receive platinum-based adjuvant chemotherapy and

460 to observation until disease progression (223). After a median follow-up of

more than 4 years, the overall survival was 82% in the chemotherapy arm and

74% in the observation arm (HR, 0.67; p = 0.001). Recurrence-free survival was

better in the chemotherapy arm: 76% versus 65% (HR, 0.64; p = 0.001). The

results of this analysis must be interpreted with caution, because most of the

patients did not undergo thorough surgical staging, but the findings suggest that

platinum-based chemotherapy should be given to patients who were not

2593optimally staged. It is generally accepted that high-risk early-stage ovarian

cancer patients, including stage I, grade 3 endometrioid, stage IC, serous and

carcinosarcoma, benefit from adjuvant chemotherapy just as advanced-stage

ovarian cancer patients do. There remains uncertainty about the impact of

chemotherapy in women with stage I clear cancers as these cancers are less

chemosensitive than high-grade serous cancers.

A randomized phase III trial of three versus six cycles of adjuvant carboplatin

and paclitaxel (CP) in 457 patients with early-stage epithelial ovarian carcinoma

was conducted by the GOG (224). An unexpectedly large number of patients (126

patients, 29%) had incomplete or inadequately documented surgical staging in

this study. The recurrence rate for six cycles was 24% lower (HR, 0.76;

confidence interval [CI], 0.5–1.13; p = 0.18) versus three cycles, but this was not

statistically significant. The estimates of probability of recurrence at 5 years were

20.1% for six cycles and 25.4% for three cycles (225). An exploratory analysis of

high-risk early-stage epithelial ovarian cancer patients from this trial reported by

Chan et al., suggested that six cycles rather than three cycles may decrease risk of

recurrence, particularly in patients with serous ovarian tumors (226).

The recommendations for therapy follow:

Patients with high-grade, high-risk stage I epithelial ovarian cancer

should be offered adjuvant chemotherapy depending on the patient’s

overall health and medical comorbidities.

Treatment with CP chemotherapy for three to six cycles is used in

these patients, whereas single-agent carboplatin may be preferable for

older women and patients with other medical comorbidities.

Advanced-Stage Epithelial Ovarian Cancer

Systemic multiagent chemotherapy is the standard treatment for metastatic

epithelial ovarian cancer (227–251). After the introduction of cisplatin in the

latter half of the 1970s, platinum-based combination chemotherapy became the

most frequently used treatment regimen in the United States. The secondgeneration platinum analogue, carboplatin, was developed to have less

toxicity than its parent compound, cisplatin (238). Fewer gastrointestinal side

effects, especially nausea and vomiting, were observed than with cisplatin, and

there was less nephrotoxicity, neurotoxicity, and ototoxicity. Carboplatin is

associated with a higher degree of myelosuppression (240). The dose of

carboplatin is calculated by using the area under the curve (AUC) and the

glomerular filtration rate (GFR) according to the Calvert formula (241). The

target AUC is 5 to 6 for previously untreated patients with ovarian cancer.

A major advance in the treatment of advanced-stage disease was the

2594incorporation of paclitaxel into the chemotherapeutic regimens in the late

1990s (224,227–230). Based on two studies (228,229), paclitaxel is included in

the primary treatment of all women with advanced-stage epithelial ovarian

cancer, unless there are contraindications to paclitaxel, such as preexisting

peripheral neuropathy.

Carboplatin and Paclitaxel

A series of randomized, prospective clinical trials with paclitaxel-containing

arms defined CP as the standard treatment protocol in advanced epithelial

ovarian cancer, although there are data to support IP chemotherapy in

selected patients (228,229,235,236). [8] Two randomized, prospective clinical

studies compared the combination of paclitaxel and carboplatin to paclitaxel and

cisplatin (235,236). In both studies, the efficacy and survivals were similar, but

the toxicity was more acceptable with the carboplatin-containing regimen (Fig.

39-15) (237).

Carboplatin and Docetaxel

Docetaxel has a different toxicity profile from paclitaxel. The SCOT-ROC

(Scottish Gynaecological Cancer Trials Group) study randomly assigned 1,077

women with stages IC to IV epithelial ovarian cancer to carboplatin with either

paclitaxel or docetaxel (244). The efficacy of docetaxel appeared to be similar

to paclitaxel: The median PFS was 15.1 months versus 15.4 months, and the

docetaxel group had fewer neurologic effects, arthralgias, myalgias, and

extremity weakness than the paclitaxel group. The docetaxel plus carboplatin

regimen was associated with significantly more myelosuppression and its

consequences (i.e., serious infections and prolonged grade 3 to 4 neutropenia).

Docetaxel is not widely used, but is an option in patients who have had a

significant allergic reaction to paclitaxel or who develop early sensory

neuropathy.

Five-Arm Trial

An Intergroup, international trial—GOG 182/Southwest Oncology Group

(SWOG) 182/ICON5—compared the standard combination of CP with these

drugs in combination with gemcitabine, topotecan, or liposomal doxorubicin in

sequential doublets or triplets (245). The study showed that the addition of any

these three drugs to the standard chemotherapy with CP did not improve the PFS

or overall survival.

Intraperitoneal Chemotherapy

A randomized, prospective GOG study (Protocol 104) of IP cisplatin versus

2595intravenous (IV) cisplatin (100 mg/m2), each given with 750 mg/m2

cyclophosphamide, was performed jointly by the SWOG and the GOG in patients

with minimal residual disease (246). [8] The IP cisplatin arm had a somewhat

longer overall median survival than the IV arm, 49 months versus 41 months

(p = 0.03). In the patients with minimal residual disease (<0.5 cm maximal

residual), paradoxically, there was no difference between the two treatments, 51

months versus 46 months (p = 0.08).

In a follow-up GOG study (Protocol 114), the dose-intense arm was initiated

by giving a moderately high dose of carboplatin (dose AUC = 9) for two

induction cycles, followed by IP cisplatin 100 mg/m2 and IV paclitaxel 135

mg/m2 over 24 hours, versus IV cisplatin 75 mg/m2 and IV paclitaxel 135 mg/m2

(247). The dose-intense arm results were slightly better—the disease progressionfree median survival was 27.6 months compared with 22.5 months for the control

arm (p = 0.02). There was no difference in overall survival (52.9 months vs. 47.6

months, p = 0.056). Based on this study, it was unclear whether dose

intensification with IP cisplatin has a sustained long-term impact on the survival

of these patients.

A third randomized prospective GOG study (Protocol 172) compared IP

cisplatin and paclitaxel versus IV cisplatin and paclitaxel (248). The combination

of cisplatin 75 mg/m2 and paclitaxel 135 mg/m2 given IV every 3 weeks was

compared with paclitaxel 135 mg/m2 IV day 1, followed by cisplatin 100 mg/m2

IP day 2, and paclitaxel 60 mg/m2 IP day 8 every 3 weeks, each given for six

cycles. Although 83% of the patients randomized to IV chemotherapy

completed all six cycles of therapy, only 42% of those treated with IP

chemotherapy completed the six cycles, principally because of catheterrelated complications. For patients in either group who could not complete

the therapy because of cisplatin-related toxicity, the chemotherapy was

switched to IV carboplatin. Comparing the IV and IP arms, the median duration

of the PFS was 18.3 and 23.8 months, respectively (p = 0.05). [8] The median

duration of the overall survival in the IV-therapy and IP-therapy groups was

49.7 and 65.6 months, respectively (p = 0.03) (248). The quality of life was

significantly worse in the IP-therapy group before cycle four and 3 to 6

weeks after treatment but not 1 year after treatment. A summary of the IPcatheter–related issues in this trial was presented (249).


7FIGURE 39-15 Survival of patients with stage III epithelial ovarian cancer treated with

carboplatin and paclitaxel versus cisplatin and paclitaxel: a Gynecologic Oncology Group

study. A: Survival by treatment. B: Survival by treatment group (micro vs. macro). (From

Ozols RF, Bundy BN, Greer BE, et al. Phase III trial of carboplatin and paclitaxel

compared with cisplatin and paclitaxel in patients with optimally resected stage III ovarian

cancer: a Gynecologic Oncology Group Study. J Clin Oncol 2003;21:3194–3200, with

permission.)

In GOG 252, 1,560 patients with stage II to IV epithelial ovarian, fallopian

tube, or peritoneal carcinoma were randomized to receive six cycles of

chemotherapy. There were three treatment arms: Arm 1, IV carboplatin AUC

6/IV weekly paclitaxel at 80 mg/m2; Arm 2, IP carboplatin AUC 6/IV weekly

paclitaxel at 80 mg/m2; Arm 3, IV paclitaxel at 135 mg/m2 on day 1/IP cisplatin

at 75 mg/m2 on day 2/IP paclitaxel 60 mg/m2 on day 8. Each arm received IV

bevacizumab at 15 mg/kg with cycles 2 through 6 of chemotherapy and then

single-agent bevacizumab for cycles 7 through 22. The trial results showed a

median PFS of 27 to 29 months in patients with optimal stage II to III disease

treated with regimens consisting of different combinations of IV and IP cisplatin,

carboplatin, and paclitaxel, in combination with bevacizumab. An analysis

limited to patients with optimal stage III tumors and no gross residual disease

found a median PFS of 31 to 34 months. By comparison, the GOG 172 trial

comparing IP and IV chemotherapy regimens in ovarian cancer had a median PFS

of 23.8 months with IP cisplatin (vs. 18.3 months with IV) with an improvement

in OS in favor of IP (252). Differences in the cisplatin arm from the GOG 172

study include a dose reduction from 100 to 75 mg and a shorter infusion time

from 24 to 3 hours. If IP is used, it would be appropriate to follow the GOG 172

protocol rather than the modified protocol with a lower dose of cisplatin.

Investigators at the Mayo Clinic reported in 2009 at the SGO that only 20% of the

105 patients they treated in the clinic could complete 3 or more cycles of the 172

protocol because of toxicity. However, the GOG 172 study did find a significant

survival advantage for IP therapy and there is still support for IP chemotherapy in

some centers.

[8] Based on these randomized trials, the IP route of administration for

carboplatin or cisplatin and paclitaxel chemotherapy in the primary

treatment of optimally resected stage III ovarian cancer is an acceptable

therapeutic alternative to IV chemotherapy with CP (250). IP chemotherapy

is best suited to patients who have optimally resected tumors, a good

performance status, and are in overall good health. Because IP

chemotherapy is more cumbersome and has a higher morbidity than IV

2598therapy, this technique of drug delivery should be individualized after

thorough discussion with the patient.

Dose-Dense Intravenous Chemotherapy

The Japanese GOG randomized 637 patients with ovarian cancer to receive

carboplatin AUC 6 and paclitaxel 180 mg/m2 every 3 weeks, or the same dose of

carboplatin and 80 mg/m2 paclitaxel every week for at least six cycles (251). The

median PFS was 28 months in the dose-dense arm and 17 months in the control

group, and the median overall survival was 100.5 months for those patients

receiving dose-dense therapy, versus 62 months for the every-3-weeks arm. They

reported relatively little difference in toxicity between the two arms and that

grade 3 to 4 neurotoxicity in particular was very low. In the 417 patients who had

stage III disease, about half had less than 1 cm maximum residual disease, similar

to the patients in GOG 172. Women with at least 1 cm of residual disease

following cytoreductive surgery appeared to benefit the most from dose-dense

therapy compared to every 3 weeks therapy. In this analysis, the median PFS was

17.6 months versus 12 months and the median overall survival was 51 months

versus 33 months, respectively. There is good evidence that weekly paclitaxel is

more effective than paclitaxel every 3 weeks in breast cancer and the

Japanese GOG study suggests that the same may apply to ovarian cancer.

Other studies have led to questioning this treatment approach in the United States.

In GOG 262, patients with stage II to IV epithelial ovarian cancer were randomly

assigned to CP every 3 weeks versus dose-dense chemotherapy (carboplatin

every 3 weeks and weekly paclitaxel). Bevacizumab administration was optional

in both arms and 84% of the patients were treated with bevacizumab. Sixty-three

percent had gross residual disease, 24% had microscopic residual disease, and in

13% the amount of residual disease was not noted.

At a median follow-up of 28 months, no difference in PFS was found between

the two groups. The subset analysis suggested a difference based on whether or

not bevacizumab was received. In patients who did not receive bevacizumab,

dose-dense treatment prolonged PFS versus the every 3-week chemotherapy

(median of 14 months vs. 10 months). Among patients treated with chemotherapy

and bevacizumab, there was no significant difference among the every 3-week

chemotherapy versus dose-dense patients (median of 15 months in both arms)

(253).

The Multicenter Italian Trial in Ovarian Cancer (MITO-7), compared every 3-

week CP to a modified dose-dense regimen where CP were delivered on a weekly

schedule (carboplatin AUC 2 and paclitaxel 60 mg/m2 on days 1, 8, 15 every 3

weeks) in stage 1C to IV epithelial ovarian cancer.

2599In this study, there was a similar PFS between the two regimens with the

weekly regimen having better quality-of-life scores. Severe myelosuppression and

neuropathy were less common in the weekly regimen (254).

[9] More convincing evidence that the dose-dense therapy may be not

superior to every 3-week chemotherapy of the JCOG study can be found in

the results from the ICON8 trial. In this trial, 1,566 predominantly European

patients were randomized to receive one of three regimens: Arm 1, carboplatin

AUC 5/6 and paclitaxel 175 mg/m2 every 3 weeks; Arm 2, carboplatin AUC 5/6

every 3 weeks and paclitaxel 80 mg/m2 weekly; Arm 3, carboplatin AUC 2 and

paclitaxel 80 mg/m2 weekly. All patients had received neoadjuvant chemotherapy

with planned interval debulking or received chemotherapy after initial primary

cytoreductive surgery. There was no benefit found for the dose-dense

regimens. The PFS was 24.4 months with every 3-week dosing, compared to

24.9 and 25.3 months in arms 2 and 3, respectively (255).

Neoadjuvant Chemotherapy

[7] Neoadjuvant chemotherapy may be considered for patients with

advanced ovarian/fallopian tube/peritoneal carcinoma who are thought to

have a low likelihood of upfront optimal cytoreduction or in women

determined to have a high perioperative risk based on a poor performance

status. A series performed by Schwartz et al. suggested that these patients treated

with neoadjuvant or cytoreductive chemotherapy had survival that was

comparable to those patients treated in the same institution with debulking

surgery followed by conventional chemotherapy (256). It is accepted that two or

three cycles of chemotherapy before cytoreductive surgery may be helpful in

patients with massive ascites and large pleural effusions. The chemotherapy may

decrease the effusions, improve the patient’s performance status, and decrease

postoperative morbidity, particularly chest morbidity (257,258).

The EORTC completed a large randomized trial in 718 patients with

advanced ovarian cancer comparing initial surgery followed by six cycles of

CP with three cycles of neoadjuvant chemotherapy followed by surgical

debulking and another three cycles of chemotherapy. The study found that the

PFS was identical in both arms (12 months) and the overall survival (30 months)

was the same in both arms (203). The morbidity of surgery was significantly less

in patients receiving neoadjuvant chemotherapy, suggesting that in selected

patients with very advanced (stages IIIC and IV) ovarian cancer and on

average had a poor performance status, two to three cycles of neoadjuvant

chemotherapy prior to surgical debulking is a reasonable option. There have

been other studies in support of neoadjuvant chemotherapy as an option for

2600advanced ovarian/ fallopian tube/peritoneal carcinoma including the CHORUS

trial. This phase III randomized, noninferiority, controlled trial was done across

87 hospitals in New Zealand and the United Kingdom and enrolled patients with

suspected stage III or IV cancer. Patients were randomly assigned to either

primary surgery followed by six cycles of chemotherapy versus three cycles of

primary chemotherapy followed by surgery, then three additional cycles of

completion chemotherapy. Patients could receive CP or another carboplatin

combination regimen or single-agent carboplatin. There was no difference in the

median overall survival comparing primary surgery to the primary chemotherapy

group. The CHORUS trial also concluded that in women with very advanced

stage III or IV ovarian cancer and a poor performance status, survival with

primary chemotherapy is noninferior to primary surgery (204).

Chemotherapy and Bevacizumab

Inhibition of angiogenesis with drugs such as bevacizumab demonstrated

activity and benefit in women with recurrent ovarian cancer. There is

evidence in other tumor types such as breast cancer and colon cancer that the

addition of bevacizumab to chemotherapy increases response rates, PFS, and

survival in some studies (259–261). Two large randomized trials (GOG 218 and

ICON7) investigating the impact of the addition of bevacizumab to standard CP

in patients with advanced ovarian cancer were carried out to address this question

(262,263). GOG 218 was a phase III three-arm randomized double-blind placebocontrolled trial. Patients in arm one received six cycles of CP and placebo starting

with the second cycle and continuing for 16 additional cycles after the completion

of chemotherapy. Patients in arm two received six cycles of chemotherapy with

bevacizumab starting with cycle two and administered with chemotherapy

followed by 16 cycles of placebo, and in arm three patients received bevacizumab

starting with cycle two of chemotherapy and then received 16 additional cycles

after the completion of chemotherapy. This study was designed to investigate the

benefit of bevacizumab in combination with chemotherapy and as a maintenance

therapy. The bevacizumab was administered at a dose of 15 mg/kg, starting with

the second cycle of chemotherapy, to decrease the risk of gastrointestinal

perforation, which is a rare complication of this agent. The GOG 218 reported a

modest improvement of 3.8 months in the PFS in patients randomized to receive

bevacizumab in combination with CP every 3 weeks and then as a maintenance

therapy every 3 weeks for an additional 16 cycles for a total of 15 months’

treatment (262). The toxicity of bevacizumab was acceptable and the risk of

bowel perforation low (below 2%). The study did not find any improvement in

the overall survival with bevacizumab in either arm receiving the drug. An

unplanned subgroup analysis demonstrated that treatment with bevacizumab

2601improved the PFS and overall survival particularly in patients with ascites but not

in other patients. The ICON7 study is similar to the GOG study, although it was a

two-arm study of CP plus or minus bevacizumab 7.5 mg/kg administered every 3

weeks with chemotherapy for six cycles and then as maintenance therapy for 12

additional cycles. The ICON7 results reported a longer median PFS at 42 months

of follow-up for the arm including bevacizumab compared to the standard therapy

arm (24 months vs. 22 months). There was no difference in the overall survival

noted (263). In an exploratory analysis of a predefined subgroup of patients with

poor prognosis/ high-risk disease, a significant difference in the overall survival

was noted between women who received bevacizumab plus chemotherapy and

those who received chemotherapy alone (restricted mean survival time, 34.5

months [95% CI, 32.0–37.0] with standard chemotherapy vs. 39.3 months [37.0–

41.7] with bevacizumab; log-rank p = 0.03). In non–high-risk patients, the

restricted mean survival time did not differ significantly between the two

treatment groups (49.7 months [95% CI, 48.3–51.1] in the standard chemotherapy

group vs. 48.4 months [47.0–49.9] in the bevacizumab group; p = 0.20) (264).

The results of these studies support a potential role of bevacizumab in

combination with chemotherapy in selected patients with advanced ovarian

cancer. However, the cost of bevacizumab is very high and the improvement

in PFS is short and no survival improvement was seen.

Chemotherapeutic Recommendation in Advanced Epithelial Ovarian Cancer

For the treatment of advanced-stage epithelial ovarian cancer, the following

treatment regimens should be considered (Table 39-4):

Combination chemotherapy or IV CP every 3 weeks or IP carboplatin

or cisplatin and paclitaxel are the treatments of choice for patients with

advanced disease. The advantages and disadvantages of the IV versus

IP routes of administration of these drugs should be discussed with the

patient.

The recommended doses and schedule for IV chemotherapy are

carboplatin (starting dose AUC = 5 to 6), and paclitaxel (175 mg/m2),

every 3 weeks for six cycles. Alternatively, the dose-dense regimen of

carboplatin AUC 6 every 3 weeks for six cycles and weekly paclitaxel 80

mg/m2 could be used, but is not superior to the 3-weekly regimen in

non-Japanese women (251). There are no good data to show any

benefit for treatment beyond six cycles but the number of cycles may

need to be individualized based on time to response and toxicity.

Ideally the CA-125 should have normalized by the end of

chemotherapy.

2602The recommended doses and schedule for IP chemotherapy are

paclitaxel 135 mg/m2 IV on day 1, followed by carboplatin AUC 5 or

cisplatin 75 to 100 mg/m2 IP on day 2, followed by paclitaxel 60 mg/m2

IP on day 8, every 3 weeks for six cycles, as tolerated.

Bevacizumab 7.5 to 15 mg/kg can be added to any of these IV or IP

chemotherapy regimens.

In patients who cannot tolerate combination chemotherapy, singleagent, IV-administered carboplatin (AUC 5 to 6) can be given.

In patients who have a hypersensitivity to paclitaxel or carboplatin, an

alternative active drug can be substituted (e.g., docetaxel, nanoparticle

paclitaxel, cisplatin). In the case of carboplatin hypersensitivity,

desensitization could be attempted.

Table 39-4 Combination Chemotherapy for Advanced Epithelial Ovarian Cancer:

Recommended Regimens

The treatment of all patients with advanced-stage disease is approached in

2603a similar manner, with modifications based on the overall status and general

health of the patient and the extent of residual disease present at the time

treatment is initiated.

Maintenance Therapy Following Response to First-Line Chemotherapy

Almost 80% of women with advanced-stage disease who respond to first-line

chemotherapy will relapse. There have been several trials conducted to determine

if there is a benefit of maintenance therapy in these patients immediately

following their primary treatment in an effort to decrease the relapse rate.

Paclitaxel

In a study conducted by the GOG and SWOG, 277 women with advanced ovarian

cancer who had a complete clinical remission to first-line chemotherapy were

randomized to receive 3 or 12 cycles of additional single-agent paclitaxel (175 or

135 mg/m2 every 28 days) (265). Patients were excluded if they developed grade

2 or 3 neurotoxicity during their initial chemotherapy. Because of cumulative

toxicity, the mean number of actual cycles of paclitaxel received by the group

assigned to receive 12 cycles was 9. The treatment-related grade 2 to 3

neuropathy was more common with longer treatment, 24% versus 14% of

patients, respectively. The study closed after a median follow-up of only 8.5

months, and an interim analysis showed a significant 7-month prolongation in the

median PFS (28 months vs. 21 months) with 9 months versus 3 months of

consolidation paclitaxel. There was no difference in the median overall survival

and this study has not changed practice. It is improbable that a survival benefit

will appear with longer follow-up, because patients assigned to three cycles were

given the option of receiving an additional nine courses of paclitaxel after the

study was discontinued (266). Another placebo-controlled, randomized trial using

two formulations of paclitaxel was conducted by the GOG. A total of 1,157

patients with stage III to IV ovarian/fallopian tube/peritoneal cancer who

achieved a clinical complete response after first-line treatment were randomized

to surveillance, paclitaxel infusion every 28 days for 12 cycles, or CT-2103

(paclitaxel poliglumex) on the same schedule. After a median follow-up of 71

months, the median overall survival was not different between the groups. There

was a small extension in the PFS with maintenance therapy compared to

surveillance (267).

Topotecan

Four additional treatment courses of topotecan were administered to patients

following six cycles of CP in two randomized trials, one conducted in Italy and

the other in Germany (268,269). In the larger trial conducted in Germany, 1,059

2604evaluable patients were randomly assigned to six cycles of paclitaxel (175 mg/m2

over 3 hours) and carboplatin (AUC 5) with (537 patients) or without (522

patients) four additional cycles of topotecan (1.25 mg/m2 IV days 1 to 5 every 3

weeks) (269). In the Italian trial, 273 women were randomly assigned to receive

four additional cycles (137 patients) of topotecan at a dose of 1 mg/m2 on days 1

to 5 every 3 weeks or no further chemotherapy (136 patients) (268). No

significant differences in either the PFS or overall survival in patients who

received four to six cycles of consolidation topotecan.

Cisplatin

In a randomized clinical trial of IP cisplatin for consolidation versus observation,

there was no difference in survival between the treatment arms (270). A metaanalysis concluded that maintenance chemotherapy was associated with increased

toxicity but no significant improvement in the PFS or overall survival (271).

Biologic Therapies

Adding the monoclonal antibody (MonAb) bevacizumab to first-line CP

chemotherapy followed by maintenance bevacizumab for a year was

associated with a modest improvement in the PFS (262,263).

There are a number of trials that addressed the role of oral angiogenesis

inhibitors as maintenance therapy after completion of first-line therapy in women

with advanced ovarian cancer. Pazopanib is an oral tyrosine kinase inhibitor of

vascular endothelial growth factor receptors, platelet-derived growth factor

receptors, and c-KIT. In the randomized double-blinded placebo-controlled phase

III AGO-OVAR16/VEG110655 trial, maintenance pazopanib for up to 2 years

after completion of first-line chemotherapy in women with advanced epithelial

ovarian cancer improved the PFS over placebo (HR, 0.77; 95% CI, 0.64–0.91; P

= 0.0021; median PFS 17.9 with PZ vs. 12.3 months with placebo) (272).

However, there was a higher incidence of grade 3 and 4 adverse events in the

pazopanib arm, which led to dose reductions in 58% of patients and early

discontinuation caused by adverse events (AE) in 33% of patients. In view of the

toxicity, the pharmaceutical company who sponsored the trial decided not to

submit for regulatory approval. Nintedanib is a triple angiokinase inhibitor of

VEGF, fibroblast growth factor, and platelet-derived growth factor. AGO-OVAR

12 randomized 1,366 patients 2:1 to maintenance nintedanib or placebo as

maintenance therapy following first-line chemotherapy. The median PFS in

nintedanib was 17.2 months vs. 16.6 months in placebo arm and this is not a

reasonable therapeutic option (273).

Studies investigating MonAbs directed toward CA-125 (OvaRex) and toward

the HMFG (human milk fat globulin) tumor–associated antigens were conducted

2605(274–276). In a randomized, placebo-controlled trial of IV oregovomab (anti-CA-

125 MonAb) as maintenance therapy, Berek et al. reported that oregovomab did

not demonstrate a survival advantage as a maintenance therapy (275). A

randomized trial of an IP-administered yttrium-labeled anti-mucin (HMFG)

MonAb versus placebo was not associated with an improved overall survival after

a negative second-look laparoscopy (276).

Treatment Assessment

Many patients who undergo optimal cytoreductive surgery and subsequent

chemotherapy for epithelial ovarian cancer have no evidence of disease at the

completion of treatment. Tumor markers and radiologic assessments are too

insensitive to exclude the presence of subclinical disease. Historically, a secondlook surgery was performed to evaluate these patients, but that was

abandoned, as there was no evidence of a meaningful benefit to patients

(161,277–286).

Tumor Markers

CA-125 is a surface glycoprotein associated with müllerian epithelial tissues, and

is elevated in about 80% of patients with epithelial ovarian cancers, particularly

those with nonmucinous tumors (287–289). The CA-125 level frequently falls

below the normal range after the initial surgical resection and one or two cycles of

chemotherapy. Carcinoembryonic antigen (CEA) levels may be elevated in

patients with ovarian cancer, but the test is too nonspecific and insensitive to be

used in the management of these patients, apart from patients with a mucinous

cancer of the ovary who may have an elevated CEA and CA19-9 (42).

Levels of CA-125 were correlated with findings at second-look operations.

Elevated levels predicted the presence of residual disease, but levels within the

normal range are an insensitive determinant of the absence of disease. In a

prospective study, the predictive value of a positive test was 100%; if the level of

CA-125 was elevated (>35 U/mL), the disease was always detectable in patients

at the second-look procedure (287). The predictive value of a negative test was

only 56%; if the level was less than 35 U/mL, disease was present in 44% of the

patients at the time of the second-look surgery. A literature review suggests that

an elevated CA-125 level predicts persistent disease at second-look surgery in

97% of the cases, but the CA-125 level is not sensitive enough to exclude

subclinical disease in many patients (288).

Serum CA-125 levels can be used during chemotherapy to follow those

patients whose levels were elevated at initial diagnosis and at the initiation of

therapy (287). The fall in the CA-125 correlates with response. Those patients

with persistently elevated levels after three cycles of treatment almost certainly

2606will still have persistent disease. The majority of patients will have a CA-125 in

the normal range following surgery and first-line chemotherapy and are typically

followed with CA-125 levels every 3 to 4 months. There are well-accepted

Gynecologic Oncology Intergroup (GCIG) criteria for CA-125 progression.

Patients with an elevated CA-125 pretreatment and in the normal range following

the completion of treatment must have a CA-125 greater than, or equal to, two

times the upper normal limit on two occasions at least 1 week apart to be

classified as having CA-125 progression (289).

Radiologic Assessment

CT can be used to assess the response in patients with measurable lesions at

the start of therapy. In patients who have no or minimal residual disease

following cytoreductive surgery, the value of these tests is limited, but they

may be useful in follow-up, especially to document the site of recurrence in

patients with CA-125 progression. Ascites can be readily detected, but other

diseases can be missed on a CT scan in patients with recurrent disease (290). A

CT scan and fine-needle aspiration (FNA) cytology, if appropriate, can diagnose

persistent or recurrent disease, but the false-negative rate of a CT scan is about

45% (291). MRI can be used as an alternative to CT in patients with allergies to

the contrast medium (131,133). PET/CT has high diagnostic accuracy in patients

with recurrent ovarian cancer. The reported sensitivity, specificity, and accuracy

range from 86% to 96%, 93% to 100%, and 85% to 100%, respectively (292).

Second-Line Therapy

Secondary Cytoreductive Surgery

Secondary cytoreduction is defined as an attempt at cytoreductive surgery in

patients with recurrent ovarian cancer. This is usually considered in patients

with platinum-sensitive recurrent ovarian cancer with localized recurrence or

limited number of sites of recurrence when it is considered possible to resect all

disease (293–296). Patients with progressive disease on chemotherapy (platinum

refractory) or with platinum-resistant ovarian cancer are not suitable candidates

for secondary cytoreduction. Although there are no hard and fast rules, secondary

debulking should be restricted to patients who have a disease-free interval of at

least 12, but preferably 24 months or those in whom it is expected that all

macroscopic disease can be resected, regardless of the disease-free interval (293–

301). The role of secondary debulking surgery has been addressed in the

DESKTOP III trial. This study included patients with a PFS OF >6 months after

first-line chemotherapy and who were considered to be good candidates for

surgery based on a positive AGO Study Group score, defined as an ECOG

performance status score of 0, ascites of 500 mL or less, and complete resection at

2607initial surgery. The PFS results have been presented at ASCO 2017 and DuBois

reported that the median PFS in 204 women who met these criteria and were

randomized to undergo surgery followed by chemotherapy was 19.6 months,

compared with 14 months in 203 women who were randomized to receive

only second-line chemotherapy. The primary endpoint of the study is overall

survival which will be available in a few years (302).

Chemotherapy for Recurrent Ovarian Cancer

The majority of women who relapse will be offered more chemotherapy with the

likelihood of benefit related to the initial response and the duration of response.

The goals of treatment include improving control of disease-related

symptoms, maintaining or improving the quality of life, delaying the time to

progression, and possibly prolonging survival, particularly in women with

platinum-sensitive recurrences. Many active chemotherapy agents (platinum,

paclitaxel, topotecan, liposomal doxorubicin, docetaxel, gemcitabine, and

etoposide) and targeted agents (bevacizumab) and PARP inhibitors are available,

and the choice of treatment is based on many factors including the likelihood of

benefit, potential toxicity, and patient convenience.

Women who relapse later than 6 months after primary chemotherapy are

classified as platinum-sensitive and usually receive further platinum-based

chemotherapy with response rates ranging from 27% to 65% and a median

survival of 12 to 24 months (303–307). Patients who relapse within 6 months

of completing first-line chemotherapy are classified as platinum-resistant and

have a median survival of 6 to 9 months and a 10% to 30% likelihood of

responding to chemotherapy. Patients who progress while on treatment are

classified as having platinum-refractory disease. Objective response rates to

chemotherapy in patients with platinum-refractory ovarian cancer are low—less

than 20% (308).

The potential adverse effects associated with chemotherapy in trials in

women with recurrent ovarian cancer are well documented and should not

be underestimated. The three most commonly used drugs are paclitaxel,

topotecan, and liposomal doxorubicin (309–335). The reported adverse effects

associated with paclitaxel include alopecia in 62% to 100%, neurotoxicity (any

grade) in 5% to 42% of patients, and grade 3 to 4 leukopenia in 4% to 24% of

patients. Topotecan is associated with significantly greater myelosuppression than

liposomal doxorubicin or paclitaxel and is observed in 49% to 76% of patients.

Liposomal doxorubicin is associated with palmar-plantar erythrodysesthesia

(PPE) of any grade in over 50% of patients and is severe in 23% particularly in

patients treated at higher doses such 50 mg/m2. Severe stomatitis is reported in up

to 10% of patients (309–312).

2608Platinum-Sensitive Disease

The use of combination platinum plus paclitaxel chemotherapy versus a singleagent platinum was tested in two multinational randomized phase III trials and a

randomized phase II study (336,337). In a report of the ICON4 and AGO-OVAR-

2.2 (AGO Studiengruppe Ovarialkarzinom) trials, 802 women with platinumsensitive ovarian cancer, who relapsed after being treatment free for at least 6 to

12 months were randomized to platinum-based chemotherapy (72% carboplatin

or cisplatin alone; 17% CAP; 4% carboplatin plus cisplatin; and 3% cisplatin

plus doxorubicin) or paclitaxel plus platinum-based chemotherapy (80%

paclitaxel plus carboplatin; 10% paclitaxel plus cisplatin; 5% paclitaxel plus

both carboplatin and cisplatin; and 4% paclitaxel alone) (336). The AGOOVAR-2.2 trial did not accrue its planned number of patients. In both trials, a

significant proportion of the patients did not receive paclitaxel as part of

their initial chemotherapeutic regimen. Combining the trials for analysis,

there was a significant survival advantage for the paclitaxel-containing

therapy (HR, 0.82) with a median follow-up of 42 months. The absolute 2-year

survival advantage was 7% (57% vs. 50%), and there was a 5-month

improvement in median survival (29 months vs. 24 months). The PFS was

better with the paclitaxel regimen (HR, 0.76); there was a 10% difference in 1-

year PFS (50% vs. 40%) and a 3-month prolongation in the median PFS (13

months vs. 10 months). The toxicities were comparable, except for a significantly

higher incidence of neurologic toxicity and alopecia in the paclitaxel group, while

myelosuppression was significantly greater with the non-paclitaxel–containing

regimens. These data support the slight advantage of a second-line regimen

containing both paclitaxel and a platinum agent compared with platinumbased therapy alone. There were two randomized trials comparing

carboplatin alone to carboplatin and gemcitabine or liposomal doxorubicin

(338,339). There was a higher response rate with the combination therapy and a

longer PFS, but the studies were not powered to look at the overall survival. In

the GCIG study comparing carboplatin and gemcitabine with carboplatin alone,

the response rate was 47.2% for the combination and 30.9% for carboplatin, with

the PFS being 8.6 months and 5.8 months, respectively (338). A large GCIG

study (CALYPSO) compared carboplatin and liposomal doxorubicin (CD)

with CP in 976 patients (340). The PFS for the CD arm was statistically

superior to the CP arm with a median PFS of 11.3 months versus 9.4 months,

respectively. There was no statistically significant difference in the overall

survival between the treatment groups. Median survival was 33 months versus

30.7 months for CP versus CD. The CD arm was better tolerated with less

severe toxicities, and this combination is now widely used (340).

Carboplatin, gemcitabine, and bevacizumab have been shown in a phase III

2609randomized control trial to be superior to carboplatin and gemcitabine alone

for platinum-sensitive recurrent epithelial ovarian cancer. In the OCEANS

study, 484 with platinum-sensitive disease were randomly assigned to carboplatin

(AUC 4 on day 1) and gemcitabine 1,000 mg/m2 on days 1 and 8 with or without

bevacizumab (15 mg/kg on day 1) with every 21-day cycles. Bevacizumab could

be given concurrently with chemotherapy for a maximum of 10 cycles followed

by bevacizumab alone until progression of disease or toxicity.

The addition of bevacizumab to carboplatin and gemcitabine resulted in an

improvement in the PFS (12 months vs. 8 months; HR, 0.48; 95% CI, 0.39–0.61);

however, there was no overall survival difference between the two arms.

Treatment with bevacizumab was associated with higher rates of serious

hypertension (17% vs. <1%), proteinuria grade 3 or higher (9% vs. 1%) and

noncentral nervous system bleeding (6% vs. 1%) (341,342).

In GOG 213, patients with platinum-sensitive recurrent epithelial ovarian

cancer were assigned randomly to secondary cytoreduction versus no secondary

cytoreduction and separately to chemotherapy (CP) with or without bevacizumab.

Patients treated with bevacizumab received the drug with chemotherapy and

afterward it was continued as a single agent until disease progression.

Compared with chemotherapy alone, the addition of bevacizumab resulted in

an improvement in the PFS (13.8 months vs. 10.4 months, respectively; HR, 0.61;

95% CI, 0.51–0.72). There was an improvement of the overall survival of 5

months with chemotherapy and bevacizumab versus chemotherapy alone; median

overall survival (42.6 months vs. 37.3 months, respectively; HR, 0.84; 95% CI,

0.69–1.01) (343). These studies show that maintenance with bevacizumab can

improve the PFS and may improve the overall survival in a subset of

patients.

PARP Inhibitors

PARP inhibitors, including niraparib and olaparib, have been approved by

FDA for maintenance therapy in patients with platinum-sensitive disease

following a partial or complete response to platinum-based chemotherapy.

The PFS is improved with these agents regardless of the BRCA status,

although the patients with BRCA germline/somatic mutations have a greater

benefit than patients without mutations.

Niraparib

Niraparib has been approved by the U.S. FDA for maintenance therapy in

patients with platinum-sensitive disease following a partial or complete

response to platinum-based chemotherapy for both germline BRCA1 and

BRCA2 carriers and nongermline carriers.

2610In the NOVA study, a phase III trial, 553 patients with platinum-sensitive

recurrent ovarian cancer were assigned randomly after completion of

platinum-based chemotherapy in a 2:1 ratio to niraparib maintenance or

placebo. Patients were stratified based on the presence or absence of a gBRCA

mutation. The patients in the non-gBRCA mutation cohort were further classified

by whether or not homologous recombination deficiency (HRD) was observed.

Niraparib compared to placebo increased the PFS in all groups of patients. In the

gBRCA group, the PFS was 21 months versus 5.5 months (HR, 0.27; 95% CI,

0.17–0.41) in contrast to the non-gBRCA group, where the PFS was 9.3

months versus 3.9 months (HR, 0.45; 95% CI, 0.34–0.61). This study also

attempted to identify the subset of patients with tumors with HRD and reported

that in the HRD-positive group of non-gBRCA patients, the PFS was 12.9 months

versus 3.8 months (HR, 0.38; 95% CI, 0.24–0.59). An exploratory analysis of

HRD-negative patients demonstrated an improvement in the median PFS in favor

of niraparib (6.9 months vs. 3.8 months; HR, 0.58; 95% CI, 0.08–0.90) (344).

Olaparib

Olaparib is FDA approved for the treatment of patients with gBRCAmutated recurrent ovarian cancer who have received three or more prior

lines of chemotherapy.

There are data in support of maintenance olaparib in women with

platinum-sensitive recurrent high-grade serous ovarian cancer from Study

19. This was a randomized phase II controlled trial of approximately 300 women

with platinum-sensitive recurrent high-grade ovarian cancer who had achieved a

response to their most recent treatment regardless of their BRCA status. The

patients were randomly assigned to olaparib 400 mg twice daily or placebo.

Compared with placebo, olaparib was shown to improve the PFS (8 months vs. 5

months; HR for progression or death, 0.35; 95% CI, 0.25–0.49) (345). A separate

analysis of Study 19 evaluated outcomes for gBRCA patients and showed that the

clinical benefit was greatest for these patients. The PFS was (11 months vs. 4

months; HR, 0.18; 95% CI, 0.10–0.31) for olaparib versus placebo, respectively

(346,347).

A randomized, double-blind, placebo-controlled, phase 3 trial was of

maintenance olaparib after first-line combination chemotherapy known as the

SOLO1 study was conducted in 15 countries (348). Of the 391 patients who

underwent randomization, 260 were assigned to receive olaparib and 131 to

receive placebo. A total of 388 patients had a germline BRCA1 or BRCA2

mutation, and 2 patients had a somatic BRCA1 or BRCA2 mutation. After a

median follow-up of 41 months, the risk of disease progression or death was 70%

lower with olaparib than with placebo (at 3 years, 60% vs. 27%, HR = 0.30; 95%

2611CI, 0.23 to 0.41; p <0.0001). Adverse events were consistent with the known

toxic effects of olaparib. The use of maintenance therapy with olaparib

provided a substantial benefit in progression-free survival among women

with newly diagnosed advanced ovarian cancer and a BRCA1 or BRCA2

mutations, and they had a 70% lower risk of disease progression or death

with olaparib than with placebo.

These results were confirmed in the much larger SOLO2 trial, which included

295 patients with gBRCA mutations and platinum-sensitive recurrent ovarian

cancer who had responded to platinum-based chemotherapy. They were randomly

assigned to receive olaparib (n = 196) or placebo (n = 99). The median PFS was

significantly longer with olaparib (19.1 months [95% CI, 16.3–25.7]) than with

placebo (5.5 months [5.2–5.8]; HR, 0.30; 95% CI, 0.22–0.41]; p <0.0001).

Rucaparib

Rucaparib is approved for treatment of BRCA-mutation–associated

advanced ovarian cancer after completion of treatment with two or more

chemotherapy regimens regardless of whether patients are platinumsensitive or resistant.

Ariel 3 included patients with platinum-sensitive, high-grade serous or

endometrioid ovarian, fallopian tube, or peritoneal carcinoma, who had received

at least two prior platinum-based chemotherapy regimens, complete or partial

response to their last platinum-based regimen, and a CA-125 concentration of less

than the upper limit of normal. They were randomized 2:1 to receive oral

rucaparib 600 mg twice daily or placebo. The median PFS in patients with a

BRCA-mutant carcinoma was 16.6 months (95% CI, 13.4–22.9; 130 [35%]

patients) in the rucaparib group versus 5.4 months (3.4–6.7; 66 [35%] patients) in

the placebo group (HR, 0.23 [95% CI, 0.16–0.34]; p <0.0001). Similar to the

NOVA trial, the investigators analyzed all tumors for HRD using a different

method. In patients with an HRD carcinoma (236 [63%] vs. 118 [62%]), the PFS

was 13.6 months (10.9–16.2) versus 5.4 months (5.1–5.6; 0.32 [0.24–0.42]; p

<0.0001).

In a phase II trial of 204 patients with recurrent platinum-sensitive, high-grade

ovarian cancer treated with rucaparib, patients with BRCA mutations and high

loss of heterozygosity (LOH) had a longer PFS compared to patients with low

LOH. Patients were divided into three groups for the study; those with germline

or somatic BRCA mutations, those who were BRCA-wild type but LOH high,

and those who were BRCA-wild type and LOH low. The PFS was longer for

patients with BRCA mutations (12.8 months; HR, 0.27; 95% CI, 0.16–0.44)

and/or LOH high (5.7 months; HR, 0.62; 95% CI, 0.42–0.90) subgroups

compared to those with LOH low (5.2 months) (349).

2612Other PARP Inhibitors

There are a number of other PARP inhibitors under investigation. Veliparib is

another PARP inhibitor with encouraging results in a phase II trial of 50 women

with BRCA mutations who were treated with veliparib monotherapy at a dose of

400 mg twice daily. There was a 26% overall response rate for platinum-sensitive

and resistant patients and a 35% response rate among those with platinumsensitive disease (350). Veliparib is being investigated in GOG-3005 in

combination with chemotherapy in the frontline and maintenance setting.

Platinum-Resistant and Refractory Disease

Patients with platinum-refractory and resistant ovarian cancer are often

treated with chemotherapy and may have a number of lines of therapy

depending on the response and performance status. In platinum-refractory

patients (i.e., those progressing on treatment), response rates to second-line

chemotherapy are less than 10% and the median survival is short, around 3

to 5 months (304–308). The management of women who are platinumresistant (i.e., progressing within 6 months of completion of chemotherapy) is

difficult and “non–cross-resistant agents” are selected, but there does not

appear to be one best treatment. Single-agent therapy is typically used because

combination regimens are associated with more toxicity without any apparent

additional benefit. High response rates of 48% to 64% were reported with dosedense weekly carboplatin (AUC 2) plus paclitaxel (80 mg/m2), and this deserves

more study (351). There are a variety of potentially active drugs: paclitaxel,

docetaxel, topotecan, liposomal doxorubicin, gemcitabine, oral etoposide,

tamoxifen, and bevacizumab are the most frequently used. Other agents include

vinorelbine and newer drugs such as trabectedin.

Chemotherapy plus bevacizumab leads to improved responses in platinumresistant disease. In the Aurelia study, 361 patients with platinum-resistant,

bevacizumab naïve ovarian cancer (progression less than or equal to 6 months

after 4 or more platinum-based cycles) were randomly assigned to treatment with

chemotherapy with or without bevacizumab at 15 mg/kg every 3 weeks. No more

than two prior lines were allowed.

Chemotherapy options included the investigator’s choice of paclitaxel 80

mg/m2 on days 1, 8, 15, and 22 every 4 weeks, or topotecan 4 mg/m2 on days 1,

8, 15 every 4 weeks or 1.25 mg/m2 on days 1 through 5 every 3 weeks, or Doxil

40 mg/m2 on day 1 every 4 weeks. Patients who received chemotherapy alone

were allowed to crossover at time of disease progression to single-agent

bevacizumab. The median follow-up was 13.5 months and compared to

chemotherapy alone, the addition of chemotherapy to bevacizumab resulted in a

2613reduction in the risk of disease progression. Median duration of response was 6.7

months versus 3.4 months (HR, 0.48; 95% CI, 0.38–0.60) in favor of

bevacizumab and chemotherapy. There was no statistical improvement in overall

survival. A planned subset analysis was performed and showed among those

patients who received paclitaxel, the median PFS with or without bevacizumab

was longer, 10 months versus 4 months, respectively; HR, 0.46; 95% CI, 0.30–

0.71 (352).

The results of a study comparing topotecan with liposomal doxorubicin

demonstrate the low response rates and poor prognosis among women with

platinum-resistant ovarian cancer (309). There were two randomized trials

comparing liposomal doxorubicin with either topotecan or paclitaxel. In a study

of 237 women who relapsed after receiving one platinum-containing regimen,

117 of whom (49.4%) had platinum-refractory disease, liposomal doxorubicin 50

mg/m2 over 1 hour every 4 weeks was compared with topotecan 1.5 mg/m2/day

for 5 days every 3 weeks (309). The two treatments had a similar overall

response rate (20% vs. 17%), time to progression (22 weeks vs. 20 weeks), and

median overall survival (66 weeks vs. 56 weeks). The myelotoxicity was

significantly lower in the liposomal doxorubicin–treated patients than with those

receiving topotecan. In a second study comparing liposomal doxorubicin with

single-agent paclitaxel in 214 platinum-treated patients who had not received

prior taxanes, the overall response rates for liposomal doxorubicin and paclitaxel

were 18% versus 22%, respectively, and median survival durations were 46 and

56 weeks, respectively, and these were not significantly different (310). In

practice, most patients are treated with a starting dose of 40 mg/m2 of liposomal

doxorubicin every 4 weeks, because of the toxicity associated with the higher

dose and the need to dose reduce when 50 mg/m2 is used. In a subset analysis of

platinum-resistant patients, the median time to progression ranged from 9.1 to

13.6 weeks for topotecan and liposomal doxorubicin, respectively. The median

survival (p = 0.455) was 35.6 weeks for pegylated liposomal doxorubicin and

41.3 weeks for topotecan. Objective response rates were recorded in 6.5% of

patients who received topotecan and in 12.3% of those who received pegylated

liposomal doxorubicin (p = 0.118). It is not known whether the treatmentimproved symptom control or quality of life because this was not specifically

addressed.

In another randomized trial of 195 patients with platinum-resistant ovarian

cancer, patients were randomized to receive either liposomal doxorubicin (PLD)

or gemcitabine (353). In the gemcitabine and PLD groups, the median PFS was

3.6 months versus 3.1 months; the median overall survival was 12.7 months

versus 13.5 months; the overall response rate was 6.1% versus 8.3%; and in the

2614subset of patients with measurable disease, the overall response rate was 9.2%

versus 11.7%, respectively. None of the efficacy end points showed a statistically

significant difference between treatment groups. The PLD group experienced

significantly more hand–foot syndrome and mucositis; the gemcitabine group

experienced significantly more constipation, nausea or vomiting, fatigue, and

neutropenia.

Taxanes

Single-agent paclitaxel shows objective responses in 20% to 30% in phase II

trials of women with platinum-resistant ovarian cancer (313–318). The main

toxicities are fatigue and peripheral neuropathy. Weekly paclitaxel appears to be

more active than 3-weekly administration in patients with platinum-resistant

ovarian cancer. In a study of 53 women with platinum-resistant ovarian cancer,

weekly paclitaxel (80 mg/m2 over 1 hour) had an objective response of 25% in

patients with measurable disease, and 27% of patients without measurable disease

had a 75% decline in serum CA-125 levels (313).

Docetaxel has some activity in these patients (354–356). The GOG studied 60

women with platinum-resistant ovarian or primary peritoneal cancer (356).

Although there was a 22% objective response rate, the median response duration

was only 2.5 months, and therapy was complicated by severe neutropenia in

three-quarters of the patients.

Topotecan

Topotecan is a relatively active second-line treatment for patients with platinumsensitive and platinum-resistant disease (321–335). In a study of 139 women

receiving topotecan 1.5 mg/m2 daily for 5 days, response rates were 19% and

13% in patients with platinum-sensitive and platinum-resistant disease,

respectively (331). The predominant toxicity of topotecan is hematologic,

especially neutropenia. With the 5-day dosing schedule, approximately 70% to

80% of patients have severe neutropenia, and 25% have febrile neutropenia with

or without infection. In some studies, regimens of 5 days produce better response

rates than regimens of shorter duration, but in others, reducing the dose to 1.0

mg/m2 per day for 3 days is associated with similar response rates but lower

toxicity (324,334). In a study of 31 patients, one-half of whom were platinum

refractory, topotecan 2 mg/m2 per day for 3 days every 21 days had a 32%

response rate (329). Continuous infusion topotecan (0.4 mg/m2 per day for 14 to

21 days) had a 27% to 35% objective response rate in platinum-refractory patients

(328). Weekly topotecan administered at a dose of 4 mg/m2 per week for 3

weeks with a week off every month produced a response rate similar to the 5-

2615day regimen with considerably less toxicity, and this dose schedule is

commonly used in the recurrent setting (334).

Oral topotecan, not available in the United States, results in similar response

rates with less hematologic toxicity (330). The IV and oral formulations of

topotecan were compared in a randomized trial of 266 women as a third-line

regimen after an initial platinum-based regimen (335). Compared with IV

topotecan (1.5 mg/m2 daily for 5 days every 3 weeks), oral topotecan (2.3 mg/m2

per day for 5 days every 3 weeks) produced a similar response rate (13% vs.

20%), less severe myelosuppression, and only a slightly shorter median survival

(51 weeks vs. 58 weeks).

Liposomal Doxorubicin

Liposomal doxorubicin (Doxil in the United States and Caelyx in Europe) has

activity in platinum- and taxane-refractory disease (309–312,339). One of the

most important side effects of liposomal doxorubicin is the hand–foot

syndrome, also known as palmar-plantar erythrodysesthesia or acral

erythema, which occurs in 20% of patients who receive 50 mg/m2 every 4

weeks (310). Most oncologists administer 40 mg/m2 and escalate only if there are

no side effects. Liposomal doxorubicin has a low rate of alopecia. In a study of 89

patients with platinum-refractory disease, including 82 paclitaxel-resistant

patients, liposomal doxorubicin (50 mg/m2 every 3 weeks) produced a response

in 17% (1 complete and 14 partial responses) (312). In another study, an objective

response of 26% was reported, although there were no responses in women who

progressed during first-line therapy (309).

Gemcitabine

Gemcitabine is associated with response rates of 20% to 50%, with 15% to 30%

in patients who are platinum-resistant (357–361). The principal toxicity is

myelosuppression.

Oral Etoposide

The most common toxicities with oral etoposide are myelosuppression and

gastrointestinal issues: grade 4 neutropenia is observed in about one-fourth of

patients, and 10% to 15% have severe nausea and vomiting (362,363). A study of

oral etoposide given for a prolonged treatment (50 mg/m2 daily for 21 days every

4 weeks) had a 27% response rate in 41 women with platinum-resistant disease, 3

of whom had durable complete responses (363). In 25 patients with platinum and

taxane-resistant disease, 8 objective responses (32%) were reported.

Hormonal Therapy

2616Tamoxifen is associated with CA-125 response rates of 15% to 20% in small

studies of patients with recurrent ovarian cancer (364–370). The efficacy of

tamoxifen was analyzed in a Cochrane review which included 623 women with

recurrent epithelial ovarian cancer across several studies. Overall, 10% achieved

an objective response, although the range within individual studies was 0% to

56%. The duration of response is short-lived for patients with high-grade ovarian

cancer (366). Aromatase inhibitors (e.g., letrozole, anastrozole, and exemestane),

which have activity in metastatic breast cancer, are being studied in relapsed

ovarian cancer (371). One of the principal advantages of this class of agents is its

very low toxicity (372).

Bevacizumab

Bevacizumab was the first targeted drug to show significant single-agent

activity in ovarian cancer. It is a humanized MonAb that targets angiogenesis by

binding to VEGF-A, thereby blocking the interaction of VEGF with its receptor.

There are a number of phase II studies reported using bevacizumab in patients

with platinum-sensitive and platinum-resistant ovarian cancer with response rates

ranging from 16% to 22% in platinum-sensitive and refractory patients (373). Up

to 40% of patients had stabilization of disease for at least 6 months. A study of

low-dose metronomic chemotherapy with 50 mg of cyclophosphamide daily and

bevacizumab 10 mg/kg IV every 2 weeks showed significant activity in a study of

70 patients with recurrent ovarian cancer (374). The primary end point was PFS at

6 months. The probability of being alive and progression free at 6 months was

56%. A partial response was achieved in 17 patients (24%). The median time to

progression and survival were 7.2 and 16.9 months, respectively. The use of

bevacizumab brings potential toxicity. The side effects of bevacizumab are well

recognized and include hypertension, fatigue, proteinuria, gastrointestinal

perforation or fistula, and uncommonly, vascular thrombosis and central nervous

system ischemia, pulmonary hypertension, and bleeding and wound-healing

complications. The most common side effects are hypertension that is grade 3 in

7% of patients but is manageable in most patients. The most concerning side

effect is bowel perforation and the study by Cannistra et al. was stopped after

recruiting 44 patients because of an 11% incidence of perforation of the bowel

(375). The bowel perforation complication could be minimized by carefully

screening patients. Simpkins et al. limited bevacizumab treatment to patients

without clinical symptoms of bowel obstruction or evidence of rectosigmoid

involvement on pelvic examination or bowel involvement on CT scan (376).

Their study included 25 patients with platinum-resistant ovarian cancer who were

heavily pretreated, and they observed a response rate of 28% without any bowel

perforations or other grade 3 or 4 toxicities. This highlights the importance of

2617patient selection.

Radiation Therapy

Whole-abdominal radiation therapy was given as a treatment for recurrent

or persistent disease but is associated with high morbidity and is not used.

The principal problem with this approach is the development of acute and chronic

intestinal morbidity. As many as 30% of patients receiving this treatment develop

intestinal obstruction, which necessitated exploratory surgery with potential

morbidity (377).

Intestinal Obstruction

Patients with epithelial ovarian cancer often develop intestinal obstruction, either

at the time of initial diagnosis or in association with recurrent disease (378–393).

Obstruction may be related to a mechanical blockage or to carcinomatous ileus.

The intestinal blockage can be corrected in most patients with obstruction at

initial diagnosis. The decision to perform an exploratory procedure to ease

intestinal obstruction in patients with recurrent disease is more difficult. For

patients whose life expectancy is very short (e.g., less than 2 months), surgical

relief of the obstruction is not indicated (378–383). In those patients with a

longer projected lifespan, features predicting a reasonable likelihood of

correcting the obstruction include young age, good nutritional status, and the

absence of rapidly accumulating ascites (379).

For most patients with recurrent ovarian cancer and intestinal obstruction,

particularly small bowel obstruction, initial management should include proper

radiographic documentation of the obstruction, hydration, correction of any

electrolyte disturbances, parenteral alimentation, and intestinal intubation. For

some patients, the obstruction may be alleviated by this conservative approach. A

preoperative upper gastrointestinal radiographic series and a barium enema will

define possible sites of obstruction.

If exploratory surgery is deemed appropriate, the type of operation to be

performed will depend on the site and the number of obstructions. Multiple sites

of obstruction are not uncommon in patients with recurrent epithelial

ovarian cancer. More than one-half of the patients have small bowel obstruction,

one-third have colonic obstruction, and one-sixth have both (380–384). If the

obstruction is principally contained in one area of the bowel (e.g., the terminal

ileum), this area can be either resected or bypassed, depending on what is easier

to accomplish safely. Intestinal bypass is less morbid than resection, and in

patients with progressive cancer, the survival time after these two operations is

the same (385–390).

If multiple obstructions are present, resection of several segments of intestine is

2618usually not indicated, and intestinal bypass and/or colostomy should be

performed. A gastrostomy may be useful in this circumstance, and this can

usually be placed percutaneously (389,392).

Surgery for bowel obstruction in patients with ovarian cancer carries an

operative mortality of about 10% and a major complication rate of about

30% (378–390). The need for multiple reanastomoses and poor nutrition

increases the morbidity, which consists primarily of sepsis and enterocutaneous

fistulae. The median survival ranges from 3 to 12 months, although about 20% of

these patients survive longer than 12 months (390–393).

Survival

The prognosis for patients with epithelial ovarian cancer is related to several

clinical variables. Survival analyses based on prognostic variables are presented

(4,65,151–155,160). Including patients at all stages, patients younger than 50

years of age have a 5-year survival rate of about 40%, compared with about 15%

for patients older than 50 years.

The 5-year survival rate for carefully and properly staged patients with

stage I disease is as high as 94%, for stage II is 73%, for stage III or IV 28%

(65). The 5-year survival rate for stage IIIA is 41%, for stage IIIB about

25%, for stage IIIC 23%, and for stage IV disease 11% (Fig. 39-16). An

analysis of the National Cancer Institute’s Surveillance, Epidemiology, and

End Results (SEER) database reveals a trend toward improved survival for

ovarian cancer in the United States. In this cohort, the survival for stage I

was 93%, for stage II 70%, for stage III 37%, and for stage IV 25% (394).

Survival of patients with borderline tumors is excellent, with stage I lesions

having a 98% 15-year survival (160). When all stages of borderline tumors are

included, the 5-year survival rate is about 86% to 90%.

Patients with stage III disease with microscopic residual disease at the start of

treatment have a 5-year survival rate of about 40% to 75%, compared with about

30% to 40% for those with optimal disease, and only 5% for those with

suboptimal disease (154,155,178). Patients whose Karnofsky index (KI) is low

(<70) have a significantly shorter survival than those with a KI greater than 70

(160).

2619NONEPITHELIAL OVARIAN CANCERS

Compared with epithelial ovarian cancers, other malignant tumors of the ovary

are uncommon. Nonepithelial malignancies of the ovary account for about

10% of all ovarian cancers (2,3,395). Nonepithelial ovarian cancers include

malignancies of germ cell origin, sex cord–stromal cell origin, metastatic

carcinomas to the ovary, and a variety of extremely rare ovarian cancers (e.g.,

sarcomas). Although there are similarities in the presentation, evaluation, and

management of these patients, the tumors have unique qualities that require a

special approach (2,395–398).


FIGURE 39-16 Survival of patients with epithelial ovarian cancer by substage. (Adapted

from Heintz APM, Odicino F, Maisonneuve P, et al. Carcinoma of the ovary. In Twentysixth annual report of the results of treatment of gynaecological cancer. Int J Gynecol

Oncol. 2006;95(suppl 1): S161–S192.)

Germ Cell Malignancies

Germ cell tumors are derived from the primordial germ cells of the ovary.

2620Their incidence is about one-tenth the incidence of malignant germ cell tumors of

the testis, so most of the advances in the management of these tumors are

extrapolations from experience with the corresponding testicular tumors.

Although malignant germ cell tumors can arise in extragonadal sites such as the

mediastinum and the retroperitoneum, most germ cell tumors arise in the gonad

from undifferentiated germ cells. The variation in the site of these cancers is

explained by the embryonic migration of the germ cells from the caudal part of

the yolk sac to the dorsal mesentery before their incorporation into the sex cords

of the developing gonads (2,4,395).

Table 39-5 Histologic Typing of Ovarian Germ Cell Tumors

1. Primitive germ cell

tumors

A. Dysgerminoma

B. Yolk sac tumor

C. Embryonal

carcinoma

D. Polyembryoma

E. Nongestational

choriocarcinoma

F. Mixed germ cell

tumor

3. Monodermal teratoma and somatic-type tumors

associated with dermoid cysts

A. Thyroid tumor

1. Struma ovarii

a. Benign

b. Malignant

B. Carcinoid

C. Neuroectodermal tumor

D. arcinoma

E. Melanocytic

F. Sarcoma

G. Sebaceous tumor

H. Pituitary-type tumor

2. Biphasic or triphasic I. Others

teratoma

A. Immature teratoma

B. Mature teratoma

1. Solid

2. Cystic

a. Dermoid cyst

b. Fetiform

teratoma

(homunculus)

Adapted from Kurman RJ, Carcangui ML, Herrington CS, et al., eds. World Health

Organization Classification of Tumours of Female Reproductive Organs. Lyon: IARC

Press; 2014.

Classification

A histologic classification of ovarian germ cell tumors is presented in Table 39-5

(4,395). Both α-fetoprotein (AFP) and human chorionic gonadotropin (hCG)

2621are secreted by some germ cell malignancies and can be clinically useful in

the diagnosis of a pelvic mass and monitoring patients after surgery.

Placental alkaline phosphatase (PLAP) and lactate dehydrogenase (LDH) are

elevated to 95% of dysgerminomas, and serial measurements of LDH may be

useful for monitoring the disease. A classification of germ cell tumors is based

on the histologic and immunocytochemical feature which include AFP and β-hCG

(Fig. 39-17) (399).



FIGURE 39-17 Relationship between types of pure malignant tumors. Germ cell tumors

and their secreted marker substances. (From Berek JS, Friedlander ML, Hacker NF.

Germ cell and nonepithelial ovarian cancer. In: Berek JS, Hacker NF. Berek & Hacker’s

Gynecologic Oncology. 6th ed. Philadelphia, PA: Wolters Kluwer; 2015:532.)

2622In this scheme, embryonal carcinoma (a cancer composed of

undifferentiated cells) synthesizes both hCG and AFP, and this lesion is the

progenitor of several other germ cell tumors (399–401). More differentiated germ

cell tumors, such as the endodermal sinus tumor, which secretes AFP, and the

choriocarcinoma, which secretes hCG, are derived from the extraembryonic

tissues; the immature teratomas derived from the embryonic cells have lost the

ability to secrete these substances. Pure germinomas do not secrete these markers.

Epidemiology

Although 20% to 25% of all benign and malignant ovarian neoplasms are of

germ cell origin, only about 3% of these tumors are malignant (2,4). Germ

cell malignancies account for fewer than 5% of all ovarian cancers in Western

countries. Germ cell malignancies represent up to 15% of ovarian cancers in

Asian and African American societies, where epithelial ovarian cancers are much

less common.

[11] In the first two decades of life, almost 70% of ovarian tumors are of

germ cell origin, and one-third of these are malignant (2,4,395). Germ cell

cancers are seen in the third decade, but thereafter they become quite rare.

Clinical Features

Symptoms

[11] In contrast to the slower-growing epithelial ovarian tumors, germ cell

malignancies grow rapidly and commonly present with acute/subacute pelvic

pain related to capsular distention, hemorrhage, or necrosis. The rapidly

enlarging pelvic mass may produce pressure symptoms on the bladder or rectum,

and menstrual irregularities may occur in menarcheal patients. Some young

patients misinterpret the early symptoms of a neoplasm as those of pregnancy,

which can lead to a delay in the diagnosis. Acute symptoms associated with

torsion or rupture of the adnexa can develop. These symptoms may be confused

with acute appendicitis. In more advanced cases, ascites may develop, and the

patient can have abdominal distention (389).

Signs

For a patient with a palpable adnexal mass, the evaluation can proceed as

outlined. Some patients with germ cell tumors will be premenarcheal and may

require examination under anesthesia. If the lesions are principally solid or a

combination of solid and cystic, as might be noted on an ultrasonographic

evaluation, a neoplasm is probable and a malignancy is possible (see Fig. 9.8

and Chapter 9). During the remainder of the physical examination, effort should

be directed to searching for signs of ascites, pleural effusion, and organomegaly.

2623Diagnosis

Adnexal masses measuring 2 cm or larger in premenarcheal girls or 8 cm or

larger in other premenopausal patients will usually require surgical

exploration. For young patients, blood tests should include serum hCG and AFP

and LDH. A CT scan of the chest is important because germ cell tumors can

metastasize to the lungs or mediastinum. A karyotype should be obtained

preoperatively for all premenarcheal girls, particularly those with

dysgerminomas, because they may arise in dysgenetic gonads (396,402). A

preoperative CT scan or MRI may document the presence and extent of

retroperitoneal lymphadenopathy or liver metastases; however, because these

patients require surgical exploration, preoperative imaging may not impact initial

surgical management. If postmenarcheal patients have predominantly cystic

lesions up to 8 cm in diameter, they may be observed or given oral contraceptives

for two menstrual cycles, which may reduce new ovarian cyst formation (403).

Dysgerminoma

[12] Dysgerminoma is the most common malignant germ cell tumor,

accounting for about 30% to 40% of all ovarian cancers of germ cell origin

(2,4,399). The tumors represent only 1% to 3% of all ovarian cancers, but they

represent as many as 5% to 10% of ovarian cancers in patients younger than 20

years. Seventy-five percent of dysgerminomas occur between the ages of 10 and

30 years, 5% occur before the age of 10 years, but they rarely occur after 50 years

of age (2,4,389). Because these malignancies occur in young women, 20% to 30%

of ovarian malignancies associated with pregnancy are dysgerminomas.

Germinomas are found in both sexes and may arise in gonadal or

extragonadal sites. The latter includes the midline structures from the pineal

gland to the mediastinum and the retroperitoneum. Histologically, they represent

abnormal proliferations of the basic germ cell. In the ovary, the germ cells are

encapsulated at birth (the primordial follicle), and the unencapsulated or free cells

die. If either of the latter processes fails, it is possible that the germ cell could free

itself of its normal control and multiply indiscriminately.

The size of dysgerminomas varies widely, but they are usually 5 to 15 cm in

diameter (2,4). The capsule is slightly bosselated, and the consistency of the cut

surface is fleshy and pale tan to gray-brown in color (Fig. 39-18).


FIGURE 39-18 Dysgerminoma of the ovary. Note that the lesion is principally solid with

some cystic areas and necrosis.

The histologic characteristics of the dysgerminoma are very distinctive.

The large round, ovoid, or polygonal cells have abundant, clear, very-pale–

staining cytoplasm, large and irregular nuclei, and prominent nucleoli (Fig.

39-19). Mitotic figures are seen in varying numbers, although they are usually

numerous. Another characteristic feature is the arrangement of the elements in

lobules and nests separated by fibrous septa, which are often extensively

infiltrated with lymphocytes, plasma cells, and granulomas with epithelioid cells

and multinucleated giant cells. When necrosis is extensive, the lesion may be

confused with tuberculosis. Dysgerminomas may contain syncytiotrophoblastic

giant cells and may be associated with precocious puberty or virilization. The

2625presence of these cells does not seem to alter prognosis (2,4). The presence of

calcifications should prompt a search for a possible underlying gonadoblastoma.

Because the dysgerminoma is a germ cell tumor and parthenogenesis

(stimulation of the basic germ cell to atypical division) is the accepted genesis for

the more immature teratomas, it is logical that these two tumors may coexist.

Choriocarcinoma, endodermal sinus tumor, and other extraembryonal lesions may

be associated with the dysgerminoma.


FIGURE 39-19 Dysgerminoma of the ovary. Primitive germ cells are divided into clusters

and lobules by fibrous septa rich in lymphocytes.

Approximately 5% of dysgerminomas are discovered in phenotypic

women with abnormal gonads (2,402,404). This malignancy can be associated

with patients who have pure gonadal dysgenesis (46,XY, bilateral streak gonads),

mixed gonadal dysgenesis (45,X/46,XY, unilateral streak gonad, contralateral

testis), and the androgen insensitivity syndrome (46,XY, testicular feminization).

For premenarcheal patients with a pelvic mass, the karyotype should be

2626determined (see Chapter 34).

For most patients with gonadal dysgenesis, dysgerminomas arise in

gonadoblastomas, which are benign ovarian tumors that are composed of germ

cells and sex cord stroma. If gonadoblastomas are left in situ in patients with

gonadal dysgenesis, more than 50% will develop into ovarian malignancies (404).

About 65% of dysgerminomas are stage I (i.e., confined to one or both

ovaries) at diagnosis (2,4,405–409). About 85% to 90% of stage I tumors are

confined to one ovary; 10% to 15% are bilateral. Dysgerminoma is the only

germ cell malignancy that has this significant rate of bilaterality. Other germ

cell tumors are rarely bilateral.

For patients whose contralateral ovary is preserved, disease can develop in 5%

to 10% of the retained gonads over the next 2 years (2). This figure includes those

not given additional therapy and patients with gonadal dysgenesis.

In the 25% of patients who are diagnosed initially with metastatic disease,

the tumor commonly spreads via the lymphatic system. It can spread

hematogenously or by direct extension through the capsule of the ovary with

exfoliation and dissemination of cells throughout the peritoneal surfaces.

Metastases to the contralateral ovary may be present when there is no other

evidence of spread. An uncommon site of metastatic disease is bone; when

metastasis to this site occurs, the lesions are principally in the lower vertebrae.

Metastases to the lungs, liver, and brain are often in patients with longstanding or

recurrent disease. Metastasis to the mediastinum and supraclavicular lymph nodes

is usually a late manifestation of disease (405,406).

Treatment

The treatment of patients with early dysgerminoma is primarily surgical,

including resection of the primary lesion and proper surgical staging.

Chemotherapy is administered to patients with metastatic disease. [12]

Because the disease principally affects girls and young women, special

consideration must be given to the preservation of fertility whenever

possible. An algorithm for the management of ovarian dysgerminoma is

presented in Figure 39-20.

Surgery

The minimal surgical operation for ovarian dysgerminoma is a unilateral

oophorectomy (407). If there is a desire to preserve fertility, as there almost

always is, the contralateral ovary, fallopian tube, and uterus should be left in

situ, even in the presence of metastatic disease, because of the sensitivity of

the tumor to chemotherapy. If fertility need not be preserved, it may be

appropriate to perform a total abdominal hysterectomy and bilateral salpingo-

2627oophorectomy for patients with advanced disease (409). For patients whose

karyotype analysis reveals a Y chromosome, both ovaries should be removed,

although the uterus may be left in situ for possible future embryo transfer (404).

Whereas cytoreductive surgery is of unproved value, bulky disease that can be

readily resected (e.g., an omental cake) should be removed during the initial

operation.


FIGURE 39-20 Management of dysgerminoma of the ovary. BEP, bleomycin, etoposide,

and cisplatin; CT, computed tomogram. (From Berek JS, Friedlander ML, Hacker NF.

2629Germ cell and nonepithelial ovarian cancer. In: Berek JS, Hacker NF. Berek & Hacker’s

Gynecologic Oncology. 6th ed. Philadelphia, PA: Wolters Kluwer; 2015:536.)

In patients in whom the neoplasm appears on inspection to be confined to

the ovary, a careful staging operation should be undertaken to determine the

presence of any occult metastatic disease. All peritoneal surfaces should be

inspected and palpated, and any suspicious lesions should be sampled for biopsy.

Unilateral pelvic lymphadenectomy and careful palpation and biopsy of enlarged

para-aortic nodes are particularly important parts of the staging. These tumors

may metastasize to the para-aortic nodes around the renal vessels. Dysgerminoma

is the only germ cell tumor that tends to be bilateral, excisional biopsy of any

suspicious masses is desirable (407–409). If a small contralateral tumor is found,

it may be possible to resect it and preserve some normal ovary.

Many patients with a dysgerminoma will have a tumor that is apparently

confined to one ovary and will be referred after unilateral salpingooophorectomy without surgical staging. Recommended options depend on

the pathologic findings, the results of imaging and tumor markers (BHCG

and LDH), the age of the patient and whether she desires fertility

preservation.

The options include (i) completion of surgical staging as stage 1

dysgerminomas do not require adjuvant chemotherapy, (ii) close surveillance

with pelvic and abdominal CT scans and tumor markers, or (iii) adjuvant

chemotherapy if capsular rupture or more advanced stage. Tumor markers

(LDH, AFP, and β-hCG) should be monitored in case occult mixed germ cell

elements are present.

Radiation Therapy

Dysgerminomas are very sensitive to radiation therapy, and doses of 2,500 to

3,500 cGy may be curative, even for gross metastatic disease. Loss of fertility is a

problem with radiation therapy, and radiation is rarely used as first-line treatment.

Chemotherapy

There is very good evidence to demonstrate the effectiveness of platinumbased chemotherapy, which is regarded as the treatment of choice (409–420).

The obvious advantage is the preservation of fertility (421).

[12] The most frequently used chemotherapeutic regimen for germ cell

tumors is BEP (bleomycin, etoposide, and cisplatin), EP (etoposide and

cisplatin), or EC (etoposide and carboplatin) (Table 39-6) (409–426).

Table 39-6 Combination Chemotherapy for Germ Cell Tumors of the Ovary

2630Regimen and Drugs Dose and Schedulea

BEP

Bleomycin 30,000 IU on days 1, 8, and 15 every 3 wks

Etoposide 100 mg/m2/day × 5 days every 3 wks

Cisplatin 20 mg/m2/day × 5 days, or 100 mg/m2/day × 1 day every 3

wks

VBP

Vinblastine 0.15 mg/kg days 1 and 2 every 3 wks

Bleomycin 15 units/m2/wk × 5; then on day 1 of course 4

Cisplatin 100 mg/m2 on day 1 every 3 wks

VAC

Vincristine 1–1.5 mg/m2 on day 1 every 4 wks

Actinomycin D 0.5 mg/day × 5 days every 4 wks

Cyclophosphamide 150 mg/m2/day × 5 days every 4 wks

aAll doses given intravenously.

The GOG studied three cycles of the EC regimen, consisting of etoposide (120

mg/m2 IV on days 1, 2, and 3 every 4 weeks) and carboplatin (400 mg/m2 IV on

day 1 every 4 weeks) for patients with completely resected ovarian

dysgerminoma, stage IB, IC, II, or III (417). The results showed a sustained

disease-free remission rate of 100%. This study was carried out many years ago

when carboplatin dose was based on surface area, but it is now based on AUC.

For patients with advanced, incompletely resected germ cell tumors, the GOG

studied cisplatin-based chemotherapy in two consecutive protocols (410,411). In

the first study, patients received four cycles of vinblastine (12 mg/m2 every 3

weeks), bleomycin (20 units/m2 IV every week for 12 weeks), and cisplatin (20

mg/m2 per day IV for 5 days every 3 weeks). Patients with persistent or

progressive disease at second-look laparotomy were treated with six cycles of

VAC (vincristine, actinomycin D, and cyclophosphamide). In the second trial,

patients received three cycles of BEP initially, followed by consolidation with

2631VAC, which was later discontinued in patients with dysgerminomas (411). The

VAC consolidation after BEP is no longer used. A total of 20 evaluable patients

with stages III and IV dysgerminoma were treated in these two protocols, and 19

were alive and free of disease after 6 to 68 months (median = 26 months).

Fourteen of these patients had a second-look laparotomy, and all findings were

negative. Another study at MD Anderson Cancer Center used BEP in 14 patients

with residual disease, and all patients were free of disease during long-term

follow-up (414). These results demonstrate that patients with advanced-stage,

incompletely resected dysgerminoma have an excellent prognosis when

treated with EP-based combination chemotherapy. [12] The standard

regimen is three to four cycles of BEP, depending on assigned risk based on

the data from testicular cancers, including advanced-stage seminomas which

are the male equivalent of dysgerminoma (425,429).

Recurrent Disease

About 75% of recurrences occur within the first year after initial treatment,

the most common sites being the peritoneal cavity and the retroperitoneal lymph

nodes (2,388,389). These patients should be treated with chemotherapy. Patients

with recurrent disease who had no therapy other than surgery should be treated

with chemotherapy as described above. If prior chemotherapy with BEP was

given, there are a number of second-line options including TIP (paclitaxel,

ifosfamide, cisplatin) or VIP (vinblastine, ifosfamide, cisplatin) (Table 35-7), and

consideration should be given to the use of high-dose chemotherapy in selected

patients. Patients with recurrent dysgerminomas following BEP should be

managed in specialized centers. Radiation therapy may be an option in very

selected subsets, with the major disadvantage being loss of fertility if pelvic and

abdominal irradiation is required.

Table 39-7 POMB-ACE Chemotherapy for Germ Cell Tumors of the Ovary

POMB

Day

1

Vincristine 1 mg/m2 IV; methotrexate 300 mg/m2 as a 12-hr infusion

Day

2

Bleomycin 15 mg as a 24-hr infusion: folinic acid rescue started at 24 hr

after the start of methotrexate in a dose of 15 mg every 12 hrs for 4 doses

Day

3

Bleomycin infusion 15 mg as a 24-hr infusion

Day Cisplatin 120 mg/m2 as a 12-hr infusion, given with hydration and 3-g

26324 magnesium sulfate supplementation

ACE

Days

1–5

Etoposide (VP16–213) 100 mg/m2 IV, days 1–5

Days

3–5

Actinomycin D 0.5-mg IV, days 3, 4, and 5

Day

5

Cyclophosphamide 500 mg/m2 IV, day 5

OMB

Day

1

Vincristine 1 mg/m2 IV; methotrexate 300 mg/m2 as a 12-hr infusion

Day

2

Bleomycin 15 mg as a 24-hr infusion; folinic acid rescue started at 24 hrs

after the start of methotrexate in a dose of 15 mg every 12 hrs for 4 doses

Day

3

Bleomycin 15 mg as a 24-hr infusion

IV, intravenous.

The sequence of treatment schedules is two courses of POMB followed by ACE. POMB is

then alternated with ACE until patients are in biochemical remission as measured by

human chorionic gonadotropin (hCG) and α-fetoprotein (AFP), placental alkaline

phosphatase (PLAP), and lactate dehydrogenase (LDH). The usual number of courses of

POMB is three to five. Following biochemical remission, patients alternate ACE with

OMB until remission has been maintained for approximately 12 weeks. The interval

between courses of treatment is kept to the minimum (usually 9 to 11 days). If delays are

caused by myelosuppression after courses of ACE, the first 2 days of etoposide are omitted

from subsequent courses of ACE.

From Newlands ES, Southall PJ, Paradinas FJ, et al. Management of ovarian germ cell

tumours. In: Williams CJ, Kaikorian JG, Green MR, et al., eds. Textbook of Uncommon

Cancer. New York: John Wiley & Sons; 1988:47, with permission.

Pregnancy

Because dysgerminomas tend to occur in young patients, they may coexist

with pregnancy. When a stage IA cancer is found, the tumor can be removed

intact and the pregnancy continued. For patients with more advanced disease,

continuation of the pregnancy depends on the gestational age of the fetus.

2633Chemotherapy can be given safely in the second and third trimesters in the same

dosages as given for the nonpregnant patient without apparent detriment to the

fetus (421).

Prognosis

For patients whose initial disease is stage IA (i.e., a unilateral encapsulated

dysgerminoma), unilateral oophorectomy alone results in a 5-year diseasefree survival rate of greater than 95% (408). The features associated with a

higher tendency to recur, include lesions larger than 10 to 15 cm in diameter, age

younger than 20 years, and a microscopic pattern that includes numerous mitoses,

anaplasia, and a medullary pattern (2,399,409–426,430–432).

Immature Teratomas

Immature teratomas contain elements that resemble tissues derived from the

embryo. Immature teratomatous elements may occur in combination with

other germ cell tumors as mixed germ cell tumors. [12] The pure immature

teratoma accounts for fewer than 1% of all ovarian cancers, but it is the

second most common germ cell malignancy and accounts for 10% to 20% of

all ovarian malignancies seen in women younger than 20 years (2). About

50% of pure immature teratomas of the ovary occur in women between the ages

of 10 and 20 years, and they rarely occur in postmenopausal women.

Pathology and Grading

Of fundamental importance in the understanding of the teratoma is

recognition of the maturation of the various elements. If maturation

continues along normal lines, it results in a mature teratoma, and the

prognosis is excellent as these are benign tumors with the rare exception of

other tumors arising from mature elements, such as squamous cell

carcinomas. Conversely, abnormal maturation of these elements can result in

an immature teratoma that has metastatic potential. Teratomas containing

immature elements, although relatively rare, are recognized more often as

pathologists are aware of the importance of differentiating them from mature

teratomas (Fig. 39-21). Among the tumors with embryonal elements, those

containing neural tissues demonstrate the ability to mature.

Semiquantification of immature neuroepithelium correlates with survival in

ovarian immature teratoma and is the basis for grading of these tumors (433–

435). Those with less than one low-power field (X 4) of immature

neuroepithelium on the slide with the greatest amount of such tissue (grade

1) have a survival of at least 95%, whereas greater amounts of immature

neuroepithelium (grades 2 and 3) appear to have a higher risk of recurrence

2634and a lower overall survival (approximately 85%) (435). This may not apply

to immature teratomas of the ovary in children because they have a good outcome

with surgery alone, regardless of the degree of immaturity (436,437). The

significant inter- and intraobserver difficulty with a three-tier system led some

authorities to recommend the two-tier grading system in use, with immature

teratomas categorized as either low grade or high grade (433). Immature ovarian

teratomas may be associated with gliomatosis peritonei, a favorable prognostic

finding if composed of completely mature tissues; molecular analyses suggest that

these glial implants are not tumor derived but rather represent teratoma-induced

metaplasia of pluripotent müllerian stem cells in the peritoneum (438,439).


FIGURE 39-21 Ovarian teratoma. This tumor contains both mature and immature neural

elements with a neural tube-like structure near its center.

Somatic malignant change in benign cystic teratomas was recorded as

occurring in 0.5% to 2% of cases, usually in patients older than 40 years of

age (433). The most common malignancy that develops in the initially benign

2635teratoma is squamous cell carcinoma. Other neoplasms were reported (e.g.,

adenocarcinomas, melanomas, which may arise from the skin or retinal anlage,

and sarcomas, including leiomyosarcomas and mixed mesodermal tumors) (2).

Carcinomas may arise from any of the epithelial elements.

Diagnosis

The preoperative evaluation and differential diagnosis of immature teratomas are

the same as for other germ cell tumors. Some of these tumors will contain

calcifications similar to those of mature teratomas, which can be detected by a

radiograph of the abdomen or by ultrasonography. Rarely they are associated with

the production of steroid hormones and can be accompanied by sexual

pseudoprecocity (395). Tumor markers are negative unless a mixed germ cell

tumor is present. Immature teratoma may contain embryonal hepatic or intestinal

differentiation, and the presence of these elements in a tumor with high AFP

levels. It is possible to have a slightly elevated AFP in a pure IT if they have

evidence of hepatic or intestinal differentiation, but high levels would be

associated with a mixed germ cell tumor.

Treatment

Surgery

In a premenopausal patient, immature teratomas are typically confined to a

single ovary, and therefore unilateral oophorectomy and fertility-sparing

surgical staging should be performed. For a postmenopausal patient, a total

abdominal hysterectomy and bilateral salpingo-oophorectomy may be

performed along with complete staging. Contralateral involvement is rare,

and routine resection or wedge biopsy of the contralateral ovary is

unnecessary (4,434,435). Any lesions on the peritoneal surfaces should be

sampled and submitted for histologic evaluation. The most frequent site of

dissemination is the peritoneum and, much less commonly, the retroperitoneal

lymph nodes. Blood-borne metastases to organ parenchyma, such as the lungs,

liver, or brain, are uncommon. When present, they are usually seen in patients

with late or recurrent disease and most often in tumors that are poorly

differentiated (i.e., grade 3).

It is unclear whether extensive debulking enhances the response to combination

chemotherapy (440–442). These are chemosensitive tumors and the cure depends

on the prompt delivery of chemotherapy. Therefore, any surgical resection that is

potentially morbid and could delay chemotherapy should be resisted.

Chemotherapy

Patients with IA, grade 1 tumors have an excellent prognosis, and no

2636adjuvant therapy is required. For patients whose tumors are stage IA, highgrade, adjuvant chemotherapy has been generally recommended, although

there is evidence to support close surveillance (412–414,430–434).

Chemotherapy is indicated for patients who have ascites, regardless of tumor

grade. The standard approach is BEP (412,413,443–449). The GOG investigated

three cycles of BEP for patients with completely resected stages I, II, and III

ovarian germ cell tumors (412,413,449). Overall, the toxicity was acceptable, and

91 of 93 patients treated were clinically free of disease. The BEP regimen,

which is the standard of care for testicular cancer, is the most appropriate

chemotherapy regimen for nondysgerminomatous germ cells tumors of the

ovary. As these tumors can progress rapidly, treatment should be initiated as soon

as possible after surgery, preferably within 7 to 10 days (450). The combined

BEP regimen is the international standard of care, usually administered for three

cycles in completely resected disease or in the adjuvant setting and four cycles for

macroscopic residual disease.

The switch from VBP to BEP was prompted by the experience in patients

with testicular cancer, in which the replacement of vinblastine with etoposide

was associated with a better therapeutic index (i.e., equivalent efficacy and

lower morbidity), especially less neurologic and gastrointestinal toxicity. The

inclusion of bleomycin is particularly important those patients with

nondysgerminomatous germ cell tumors. In a randomized study of three cycles

of EP with or without bleomycin (EP vs. BEP) in 166 patients with germ cell

tumors of the testes, the BEP regimen had a relapse-free survival rate of 84%

compared with 69% for the EP regimen (p = 0.03) (425). Cisplatin appears to be

superior to carboplatin in the setting of metastatic germ cell tumors. One hundred

ninety-two patients with germ cell tumors of the testes were entered into a study

of four cycles of EP versus four cycles of EC. There were three relapses with the

EP regimen versus seven with the EC regimen, although the overall survival of

the two groups is identical (426). In view of these results, BEP is the preferred

treatment regimen for patients with ovarian germ cell tumors.

It is unclear whether adjuvant chemotherapy is indicated or required for all

patients with resected immature teratomas. Several reports support the successful

management of these patients with surgery alone and close surveillance

(442,451). In the largest series, an Intergroup study from the Pediatric Oncology

Group and the Children’s Cancer Group, 73 children with immature teratoma (44

of ovarian origin) underwent surgery followed by surveillance. With a median

follow-up of 35 months, the overall 3-year event-free survival rates for all patients

and those with ovarian teratomas were 93% and 100%, respectively. Thirteen of

the 44 girls with an immature ovarian teratoma had microscopic foci of yolk sac

tumor in the teratoma; one developed recurrent disease and was successfully

2637treated with cisplatin-based chemotherapy. Eighty-two percent of the tumors were

grade 1 or 2; however, 92% of those with foci of yolk sac tumor were grade 2 or

3.

Second-Look Laparotomy or Laparoscopy

Second-look operation is not justified in patients who received adjuvant

chemotherapy, because chemotherapy in these patients is effective (431,432). In

the absence of any residual disease, there is no indication for second-look

laparotomy or laparoscopy. In patients with radiographic evidence of residual

tumor after surgery and chemotherapy, this may represent residual germ cell

tumor, benign teratoma or more commonly, necrotic tissue in a patient whose

tumor markers have normalized.

Second-look laparotomy or laparoscopy in patients with macroscopic residual

disease at the start of chemotherapy should be considered, as patients with

residual mature teratoma and are at risk of growing teratoma syndrome, an

uncommon complication of immature teratomas (452,453). Cancers can arise at a

later date in residual mature teratoma. It is important to resect or biopsy any

residual mass and exclude persistent disease because further chemotherapy may

be indicated. The principles of surgery are based on the much larger experience of

surgery in males with residual masses following chemotherapy for germ cell

tumors with a component of immature teratoma (454). Mathew et al. reported

their experience of laparotomy in assessing the nature of postchemotherapy

residue in ovarian germ cell tumors. Sixty-eight patients completed combination

chemotherapy with cisplatin regimes, and 35 had radiologic residual masses.

Twenty-nine of these 35 patients underwent laparotomy and 3 patients had viable

tumor, 7 immature teratomas, 3 mature teratoma, and 16 necrosis or fibrosis.

None of the patients with dysgerminoma, embryonal carcinoma, absence of

teratoma element in the primary tumor, and radiologic residual mass of less than 5

cm had viable tumor, whereas all patients with tumors containing the teratoma

component initially had residual tumor, strengthening the case for surgery in

patients with immature teratoma and any residual mass (455,456).

Prognosis

The most important prognostic feature of the immature teratoma is the

grade (2,423). The stage of disease and the extent of tumor at the initiation of

treatment have an impact on the curability of the lesion. Overall, the 5-year

survival rate for patients with all stages of pure immature teratomas is 70% to

80%, and it is 90% to 95% for patients with surgically staged, stage I lesions

(430,433,457).

Endodermal Sinus Tumors

2638[12] Endodermal sinus tumors (EST) are also referred to as yolk sac

carcinomas because they are derived from the primitive yolk sac (2,4). They

are the third most frequent malignant germ cell tumors of the ovary. ESTs

occur in patients with a median age of 16 to 18 years (2,4,458). About one-third

of the patients are premenarcheal at the time of diagnosis. Abdominal or pelvic

pain is the most frequent initial symptom, occurring in about 75% of patients,

whereas an asymptomatic pelvic mass is documented in 10% of patients (396).

Pathology

The gross appearance of an EST is soft grayish-brown. Cystic areas caused by

degeneration or necrosis are present in these rapidly growing lesions. The capsule

is intact in most cases.

The EST is unilateral in 100% of cases; thus, biopsy of the opposite ovary

is contraindicated. The association of such lesions with gonadal dysgenesis must

be appreciated, and chromosomal analysis should be performed preoperatively in

premenarcheal patients (4).

Microscopically, the characteristic feature is the endodermal sinus, or the

Schiller–Duval body (Fig. 39-22). The cystic space is lined with a layer of

flattened or irregular endothelium into which projects a glomerulus-like tuft with

a central vascular core. These structures vary throughout the tumor, and the

reticular, myxoid elements simulate undifferentiated mesoblast. The lining of the

papillary infolding and the cavity is irregular, with an occasional cell containing

clear, glassy cytoplasm, simulating the hobnail appearance of the epithelium in

clear cell tumors. There is a possible association of EST with dysgerminoma in

mixed germ cell tumors (2,4).


FIGURE 39-22 Yolk sac tumor of the ovary. Note the classic Schiller–Duval body with its

central vessel and mantle of endoderm.

Most EST secrete AFP and, rarely, alpha-1 antitrypsin (AAT). AFP can be

demonstrated in the tumor by immunohistochemistry. There is a good correlation

between the extent of disease and the level of AFP, although discordance can

occur. The serum level of these markers, particularly AFP, is useful in monitoring

the patient’s response to treatment (458–462).

Treatment

Surgery

The treatment of the EST consists of surgical exploration, unilateral

salpingo-oophorectomy, and a frozen section for diagnosis. The addition of a

hysterectomy and contralateral salpingo-oophorectomy does not alter the

outcome and is not indicated (460). Any gross metastases should be removed, if

possible, but thorough surgical staging may be omitted because all patients need

2640chemotherapy. At surgery, the tumors tend to be solid and large, ranging in size

from 7 to 28 cm (median, 15 cm) in the GOG series (449). Bilaterality does not

occur, and the other ovary is involved with metastatic disease only when there are

other metastases in the peritoneal cavity. Most patients have early-stage disease:

71%, stage I; 6%, stage II; and 23%, stage III (462).

Chemotherapy

All patients with ESTs are treated with either adjuvant or therapeutic

chemotherapy. Before the routine use of combination chemotherapy for this

disease, the 2-year survival rate was only about 25%. The introduction of the VAC

regimen, improved survival rates to 60% to 70%, indicating the chemosensitivity

of most of these tumors (444,445). With conservative surgery and adjuvant

chemotherapy, fertility can be preserved as with other germ cell tumors.

Cisplatin-containing combination chemotherapy with three to four cycles

of BEP should be used as primary chemotherapy for EST. The GOG

protocols used three to four treatment cycles given every 3 weeks (449,463). The

number of cycles is dependent on prognostic factors including whether there is

bulky residual disease after surgery and the site of metastases.

Rare Germ Cell Tumors of the Ovary

Embryonal Carcinoma

[12] Embryonal carcinoma of the ovary is an extremely rare tumor that is

distinguished from a choriocarcinoma of the ovary by the absence of

syncytiotrophoblastic and cytotrophoblastic cells. The patients are very young;

ages ranged between 4 and 28 years (median, 14 years) in two series (464). Older

patients were reported (465). Embryonal carcinomas may secrete estrogen, with

the patient exhibiting symptoms and signs of precocious pseudopuberty or

irregular bleeding (2). The clinical picture is otherwise similar to that of the EST.

The primary lesions tend to be large, and about two-thirds are confined to one

ovary at the time of diagnosis. These lesions frequently secrete AFP and hCG,

which are useful for following the response to subsequent therapy (461). The

treatment of embryonal carcinomas is the same as for the EST (i.e., a

unilateral oophorectomy followed by combination chemotherapy with BEP)

(413,449).

Choriocarcinoma of the Ovary

Pure nongestational choriocarcinoma of the ovary is an extremely rare tumor.

Histologically, it has the same appearance as gestational choriocarcinoma

metastatic to the ovaries (466). Most patients with this cancer are younger than 20

years. The presence of hCG can be useful in monitoring the patient’s response to

2641treatment. In the presence of high hCG levels, isosexual precocity occurs in

about 50% of patients whose lesions appear before menarche (466,467).

There are only limited reports on the use of chemotherapy for nongestational

choriocarcinomas, but complete responses were reported with the MAC

(methotrexate, actinomycin D, and cyclophosphamide) regimen used in a manner

described for gestational trophoblastic disease (see Chapter 41) (435).

Alternatively, the BEP regimen can be used. The prognosis of ovarian

choriocarcinomas is poor, with most patients having metastases to organ

parenchyma at the time of diagnosis.

Polyembryoma

Polyembryoma of the ovary is another extremely rare tumor, which is composed

of embryoid bodies. This tumor replicates the structures of early embryonic

differentiation (i.e., the three somatic layers: endoderm, mesoderm, and ectoderm)

(2,399). They tend to occur in very young, premenarcheal girls with signs of

pseudopuberty and elevated AFP and hCG levels. The same principles of

management apply (399,468).

Mixed Germ Cell Tumors

Mixed germ cell malignancies of the ovary contain two or more elements of

the lesions described above. In one series, the most common component of a

mixed malignancy was dysgerminoma, which occurred in 80%, followed by EST

in 70%, immature teratoma in 53%, choriocarcinoma in 20%, and embryonal

carcinoma in 16% (468). The most frequent combination is a dysgerminoma

and an EST. The mixed germ cell tumors may secrete either AFP, hCG,

both, or neither of these markers, depending on the components.

These tumors should be managed with BEP combination chemotherapy. The

serum marker, if positive initially, may become negative during chemotherapy,

but this finding may reflect regression of only a particular component of the

mixed lesion.

The most important prognostic features are the size of the primary tumor

and the relative size of its most malignant component (469). For stage IA

tumors smaller than 10 cm, survival is 100%. Tumors composed of less than onethird EST, choriocarcinoma, or grade 3, immature teratoma have an excellent

prognosis, but it is less favorable when these components constitute most of the

mixed lesions.

Late Effects of Treatment of Malignant Germ Cell Tumors of the Ovary

Although there are substantial data regarding late effects of cisplatin-based

therapy in men with testicular cancer, there is much less information available for

2642women with ovarian germ cell tumors. However, it is likely that the late effects

will be similar to men treated with BEP and include renal and gonadal

dysfunction, neurotoxicity, cardiovascular toxicity, and secondary malignancies.

Gonadal Function

An important cause of infertility in patients with ovarian germ cell tumors is

unnecessary bilateral salpingo-oophorectomy and hysterectomy. Although

temporary ovarian dysfunction or failure is common with platinum-based

chemotherapy, most women will resume normal ovarian function, and

childbearing is usually preserved (402,408). In one representative series of 47

patients treated with combination chemotherapy for germ cell malignancies,

91.5% resumed normal menstrual function, and there were 14 healthy live births

and no birth defects (423). Factors such as older age at the initiation of

chemotherapy, greater cumulative drug dose, and longer duration of therapy all

have an adverse effect on future gonadal function.

Secondary Malignancies

An important cause of late morbidity and mortality in patients receiving

chemotherapy for germ cell tumors is the development of secondary tumors.

Etoposide in particular was implicated in the development of treatmentrelated leukemias (470,471).

The chance of developing treatment-related leukemia following etoposide is

dose related. The incidence of leukemia is approximately 0.4% to 0.5%

(representing a 30-fold increased likelihood) in patients receiving a cumulative

etoposide dose of less than 2,000 mg/m2, compared with as much as 5%

(representing a 336-fold increased likelihood) in those receiving more than 2,000

mg/m2 (470). In a typical three- or four-cycle course of BEP, patients receive a

cumulative etoposide dose of 1,500 or 2,000 mg/m2, respectively.

Despite the risk of secondary leukemia, risk–benefit analyses concluded that

etoposide-containing chemotherapy regimens are beneficial in advanced germ cell

tumors; one case of treatment-induced leukemia would be expected for every 20

additionally cured patients who receive BEP as compared with platinum,

vincristine, bleomycin (PVB). The risk–benefit balance for low-risk disease, or for

high-dose etoposide in the recurrence setting, is less clear (471–473).

Surveillance Versus Immediate Adjuvant Chemotherapy in Stage I Germ Cell Tumors

Close surveillance is a well-accepted and standard approach to the

management of young men with apparent stage I testicular germ cell tumors.

There is a large body of evidence to support this approach, and guidelines on

surveillance protocols (474). Although as many as 20% to 30% of patients will

2643relapse, almost all will be cured using second-line chemotherapy with BEP,

and the potential adverse effects of chemotherapy can therefore be avoided in

most patients. Although this is a very common approach to management of young

men with stage I testicular germ cell tumors, it has not been widely adopted in

females with ovarian germ cell tumors in the United States. In the United

Kingdom, all women with stage IA malignant ovarian germ cell tumors are

offered surveillance regardless of histologic subtype. However, prior to making

this recommendation there is close scrutiny of operative findings, central

pathologic review, measurement of tumor markers, and whole-body imaging to

ensure that the staging is correct. Surveillance includes regular clinical review,

physical examination, interval imaging, and tumor marker assessments (hCG,

AFP, and LDH). The frequency of surveillance with tumor marker monitoring is

highest early on, as most relapses occur within 2 years.

The Charing Cross Group initially reported a prospective study of 24

patients with stage IA ovarian germ cell tumors who were also enrolled in a

surveillance program. The group consisted of nine patients (37.5%) with

dysgerminoma, nine (37.5%) with pure immature teratoma, and six (25%) with

EST (with or without immature teratoma). Treatment consisted of surgical

resection without adjuvant chemotherapy, followed by a surveillance program of

clinical, serologic, and radiologic review. A second-look operation was

performed, and all patients, but one, were alive and in remission after a

median follow-up of 6.8 years. The 5-year overall survival was 95%, and the 5-

year disease-free survival was 68%. Eight patients required chemotherapy for

recurrent disease or second primary ovarian germ cell tumor. This included three

patients with grade II immature teratoma, three patients with dysgerminoma, and

two patients with dysgerminoma who developed a contralateral dysgerminoma

4.5 and 5.2 years after their first tumor. All but one, who died of a pulmonary

embolus, were successfully salvaged with chemotherapy. The same group

updated its experience and reported on the safety of the ongoing surveillance

program of all stage IA female germ cell tumors (474). Thirty-seven patients

(median age 26, range 14 to 48 years) with stage I disease were referred to Mount

Vernon and Charing Cross Hospitals between 1981 and 2003. Patients underwent

surgery and staging followed by intense surveillance, which included regular

tumor markers and imaging. The median period of follow-up was 6 years.

Relapse rates for stage IA nondysgerminomatous tumors and

dysgerminomas were 8 of 22 (36%) and 2 of 9 (22%), respectively. In

addition, one patient with mature teratoma and glial implants relapsed. Ten of

these 11 patients (91%) were successfully cured with platinum-based

chemotherapy. Only one patient died from chemoresistant disease. All relapses

occurred within 13 months of initial surgery. The overall disease-specific

2644survival of malignant ovarian germ cell tumors was 94%. If this approach is

considered, then the Charing Cross surveillance protocol should be followed.

Sex Cord–Stromal Tumors

Sex cord–stromal tumors of the ovary account for about 5% to 8% of all

ovarian malignancies (2,4,395,396,475–481). This group of ovarian neoplasms

is derived from the sex cords and the ovarian stroma or mesenchyme. The tumors

usually are composed of various combinations of elements, including the

“female” cells (i.e., granulosa and theca cells) and “male” cells (i.e., Sertoli and

Leydig cells), and morphologically indifferent cells. A classification of this group

of tumors is presented in Table 39-8.

Granulosa–Stromal Cell Tumors

[13] Granulosa cell tumors are the most common type of malignant ovarian

sex cord–stromal tumor and make up 2% to 5% of all ovarian malignancies

Granulosa–stromal cell tumors include granulosa cell tumors, thecomas, and

fibromas. They account for 70% of ovarian sex cord–stromal tumors.

Granulosa cell tumors are considered to be a low-grade malignancy.

The majority of granulosa cell tumors are adult (95%) with juvenile granulosa

cell tumors being much less common (5%). Adult granulosa cell tumors almost

always occur in postmenopausal women while juvenile granulosa cell tumors are

diagnosed in women under 30 years of age and may be seen in prepubertal girls in

5% of cases (478). [13] Granulosa cell tumors are bilateral in only 2% of patients

(482).

Table 39-8 Sex Cord–Stromal and Steroid Cell Tumors

1. Granulosa-stromal cell tumors

A. Granulosa cell tumor

B. Tumors in thecoma–fibroma group

a. Thecoma

b. Fibroma

b. Unclassified

2. Androblastomas; Sertoli–Leydig cell tumors

A. Well differentiated

2645a. Sertoli cell tumor

b. Sertoli–Leydig cell tumor

c. Leydig cell tumor; hilus cell tumor

B. Moderately differentiated

C. Poorly differentiated (sarcomatoid)

D. With heterologous elements

3. Gynandroblastoma

4. Sex cord tumor with annular tubules

5. Sex cord–stromal tumors, unclassified

6. Steroid cell tumors

A. Stromal luteoma

B. Leydig cell tumor

C. Steroid cell tumor, not otherwise classified

Pathology

Granulosa cell tumors range from a few millimeters to 20 cm or more in diameter.

The tumors are rarely bilateral and have a smooth, lobulated surface. The solid

portions of the tumor are granular, frequently trabeculated, and yellow or grayyellow in color. After clear cell carcinoma, the granulosa-theca cell tumor is

probably the most inaccurately diagnosed tumor of the female gonad. Of 477

ovarian tumors from the Emil Novak Ovarian Tumor Registry diagnosed initially

as granulosa-theca cell tumors, almost 15% were reclassified after histologic

review. The FOXL2 gene is mutated in almost all adult granulosa cell tumors. It

plays a role in the development of normal granulosa cells and mutations in this

gene have been found in up to 97% of adult granulomas cell tumors. Molecular

testing for mutations in the FOXL2 gene may help to improve the diagnostic

accuracy among patients with sex cord–stromal tumors (483,484). Tumors

misdiagnosed as granulosa cell tumors included primary or metastatic

carcinomas, teratoid tumors, and poorly differentiated mesothelial tumors (475).

A Norwegian study found that almost 30% of adult granulosa cell tumors had

been misdiagnosed after reviewing the pathology and testing for FOXL2

mutations, which raises questions about the reliability of historical series.

2646Juvenile granulosa cell tumors, so named because of their tendency to occur in

younger patients, feature rounder, more hyperchromatic nuclei, and may contain

numerous mitotic figures. The presence of large, irregular follicle spaces is an

additional distinguishing feature of the juvenile granulosa cell tumor.

The classic adult granulosa cell is round or ovoid with scant cytoplasm. The

nucleus contains compact, finely granular chromatin and is either euchromatic or

hypochromatic (4). Better-differentiated granulosa cell tumors may have a variety

of patterns including macrofollicular, microfollicular, trabecular, solid-trabecular,

and insular while less-differentiated tumors have a more diffuse pattern described

as sarcomatoid. “Coffee bean” grooved nuclei are characteristic; mitotic figures

may be present, but numerous mitotic figures should prompt consideration for

poorly differentiated or undifferentiated carcinoma. In the most common

variety, the adult granulosa cells show a tendency to arrange themselves in

small clusters or rosettes around a central cavity, so there is a resemblance to

primordial follicles (i.e., Call–Exner bodies) (Fig. 39-23). The stroma is similar

to the theca and may be luteinized. In children and adolescents, the granulosa cell

tumors are often cystic, contain luteinized cells, and can be associated with

precocious puberty. The adult granulosa cell tumor tends to occur in older women

and the juvenile tumor in children and young women, with the diagnosis not

being based on age of presentation, but on histology. Adult granulosa cell tumors,

but not juvenile granulose cell tumors, harbor a somatic mutation in the FOXL2

gene (483).


FIGURE 39-23 Granulosa cell tumor of the ovary. Note the classic Call–Exner bodies

with a minimal stromal component in this tumor of folliculoid pattern (arrow points to an

example).

Diagnosis

About 70% of granulosa cell tumors secrete estrogen or androgens.

Inhibin is secreted by some adult granulosa cell tumors and is a useful

marker for the disease (485–489). Inhibin is a peptide hormone that is produced

by granulosa cells and plays a role in regulation of FSH secretion. Inhibin exists

in two different isoforms: inhibin A and inhibin B. Although some

laboratories only perform testing for inhibin A, serum levels of inhibin B are

more frequently elevated (490).

Inhibin levels can be used to monitor patients during follow-up or to assess

response to treatment. The lead-time between inhibin levels rising and

documented recurrence is about 12 months.

An elevated serum inhibin level in a premenopausal woman presenting with

2648amenorrhea and infertility is suggestive of a granulosa cell tumor (491). Seventyfive percent of women with AGCT’s also have an elevated anti-müllerian

hormone preoperatively and AMH has a >90% sensitivity in detecting recurrence.

Juvenile Granulosa Cell Tumors

Juvenile granulosa cell tumors of the ovary are rare and constitute less than

5% of ovarian tumors in childhood and adolescence (492). Most juvenile

granulosa cell tumors are clinically benign; only about 10% recur and when they

do, it is generally within 5 years of the initial diagnosis. Of the rare prepubertal

juvenile GCTs, 75% are associated with sexual pseudoprecocity because of

the estrogen secretion (477).

About 90% are diagnosed in stage I, and they have a favorable prognosis. The

juvenile subtype behaves less aggressively than the adult type. Only about 10% of

juvenile granulosa cell tumors are malignant and late relapse is unusual.

Advanced-stage tumors can be treated with platinum-based combination

chemotherapy (e.g., BEP) (493,494).

Adult Granulosa Cell Tumors

Low-grade endometrial cancer occurs in association with adult granulosa

cell tumors in at least 5% of cases, and 25% to 50% are associated with

endometrial hyperplasia (2,466,475–478,483). Most reproductive-age patients

have menstrual irregularities or secondary amenorrhea, and, frequently, cystic

hyperplasia of the endometrium. Abnormal uterine bleeding is often the initial

symptom for postmenopausal women. The estrogen secretion in these patients can

be sufficient to stimulate the development of endometrial cancer.

The other symptoms and signs of granulosa cell tumors are nonspecific and the

same as most ovarian malignancies. Ascites is present in about 10% of cases, and,

rarely, a pleural effusion is present (475–478). Granulosa tumors tend to be

hemorrhagic; occasionally, they rupture and produce a hemoperitoneum.

Adult granulosa cell tumors are stage I at diagnosis in 90% of patients and

are associated with a very good prognosis, but may recur 5 to 30 years after

initial diagnosis (477). The tumors may spread hematogenously, and metastases

can develop in the lungs, liver, and brain years after initial diagnosis. Malignant

thecomas are extremely rare, and their signs and symptoms, management, and

outcome are similar to those of the granulosa cell tumors (475).

Treatment

The treatment of granulosa cell tumors depends on the age of the patient and the

extent of disease. For most patients, surgery alone is sufficient primary therapy;

radiation and chemotherapy are reserved for the treatment of recurrent or

2649metastatic disease (478–481,483,485–487,492,495–497).

Surgery

Because granulosa cell tumors are bilateral in only about 2% of patients, a

unilateral salpingo-oophorectomy is appropriate therapy for stage IA tumors in

children or in women of reproductive age (476). At the time of laparotomy or

laparoscopy, if a granulosa cell tumor is identified by frozen section, a staging

operation is performed, including an assessment of the contralateral ovary. If the

opposite ovary appears enlarged, it should be sampled for biopsy. For

perimenopausal and postmenopausal women for whom ovarian preservation is

not important, a hysterectomy and bilateral salpingo-oophorectomy should be

performed. For premenopausal patients in whom the uterus is left in situ, an

endometrial biopsy should be performed because of the possibility of a

coexistent adenocarcinoma of the endometrium (478).

Radiation Therapy

There is no evidence to support the use of adjuvant radiation therapy for

granulosa cell tumors, although pelvic irradiation may help to palliate isolated

pelvic recurrences (478,495).

Chemotherapy

[13] There is no evidence that adjuvant chemotherapy will prevent

recurrence of disease and improve prognosis (497–500). Patients with

metastatic disease at initial presentation or with recurrent disease may benefit

from chemotherapy. Although the data are inconclusive, BEP or CP are the most

commonly used regimens in selected patients with stage III or IV tumors or

recurrent disease (493). In a GOG study, 37% (14 of 30) patients treated with

BEP had a negative second-look laparotomy, and these patients had a median

time to progression of 24.4 months (494). The GOG is performing a randomized

phase II trial comparing BEP with the combination of paclitaxel and carboplatin

for patients with newly diagnosed and chemo-naïve recurrent/metastatic sex cord–

stromal tumors of the ovary. Taxanes and bevacizumab are active in AGCT’s.

The Alienor trial has recently completed recruitment and randomized patients

with recurrent/metastatic sex cord–stromal tumors to either paclitaxel alone or in

combination with bevacizumab and the results are eagerly awaited. Granulosa cell

tumors are potentially hormonally responsive, with about 30% of granulosa

tumors expressing estrogen receptors and almost 100% expressing progesterone

receptors. Hormonal agents such as progestins or luteinizing hormone–releasing

hormone agonists or aromatase inhibitors have been used to treat these patients

(501–505). There are case reports of durable response to aromatase inhibitors in

patients with metastatic granulosa cell tumors who received multiple prior

2650treatment (503). Small clinical series and case reports indicated that luteinizing

hormone– releasing hormone agonists had a 50% response rate in 13 patients,

while 4 of 5 patients in the literature responded to a progestational agent. Two

case series reported durable responses in a total of 6 patients receiving aromatase

inhibitors among those who progressed on or were intolerant of chemotherapy

(502–505). There are fewer than 100 patients reported in the literature who have

received hormonal therapies and typically these have been individual case reports

or small retrospective series that are subject to bias. The role of hormonal therapy

remains uncertain.

Adult granulosa cell tumors have a prolonged natural history and a

tendency toward late relapse, reflecting their low-grade biology. Ten-year

survival rates of about 90% are reported, with 20-year survival rates dropping to

75% (476–478,491,493). Most histologic types have the same prognosis, but

patients with the more poorly differentiated diffuse or sarcomatoid type tend to do

worse (475).

The DNA ploidy of the tumors is correlated with survival. Holland et al.

reported DNA aneuploidy in 13 of 37 patients (35%) with primary adult

granulosa cell tumors (498). The presence of residual disease is the most

important predictor of PFS, but DNA ploidy is an independent prognostic factor.

Patients with residual-negative DNA diploid tumors had a 10-year PFS of

96%.

Sertoli–Leydig Tumors

Sertoli–Leydig tumors occur most frequently in the third and fourth decades

of life; 75% of these tumors are seen in women younger than 40 years. These

neoplasms are extremely rare and account for less than 0.2% of ovarian cancers

(2,506,507). Sertoli–Leydig cell tumors are most frequently low-grade

malignancies; a poorly differentiated variety may behave more aggressively (Fig.

39-24) (506–508).

The tumors typically produce androgens, and clinical virilization is noted

in 70% to 85% of patients (508). Signs of virilization include oligomenorrhea

followed by amenorrhea, breast atrophy, acne, hirsutism, clitoromegaly,

deepening of the voice, and a receding hairline. Measurement of plasma

androgens may reveal elevated testosterone and androstenedione, with normal or

slightly elevated dehydroepiandrosterone sulfate (2,509). Rarely, the Sertoli–

Leydig tumor is associated with manifestations of estrogenization (i.e., isosexual

precocity, irregular or postmenopausal bleeding). Sertoli–Leydig tumors are

associated with somatic mutations in the DICER1 gene (510).


FIGURE 39-24 Sertoli–Leydig cell tumor of the ovary. Note the aggregates of

eosinophilic Leydig cells in the stroma adjacent to Sertoli cell tubules.

Treatment

Because these low-grade lesions are rarely bilateral (<1%), the usual

treatment is unilateral salpingo-oophorectomy and evaluation of the

contralateral ovary for patients who are in their reproductive years (4,478).

For older patients, hysterectomy and bilateral salpingo-oophorectomy are

appropriate (506,507).

There are insufficient data to document the utility of radiation or chemotherapy

for patients with persistent disease, but some responses in patients with

measurable disease were reported with pelvic irradiation and the VAC

chemotherapy regimen (4,511–515).

Prognosis

The 5-year survival rate is 70% to 90%, and recurrences thereafter are

uncommon (4,509,511–515). Most fatalities occur in the presence of poorly

differentiated tumors.

2652Uncommon Ovarian Cancers

There are several varieties of malignant ovarian tumors that together constitute

only 0.1% of ovarian malignancies (2). Two of these lesions are the lipoid (or

lipid) cell tumors and the primary ovarian sarcomas.

Steroid Cell Tumors

Steroid cell tumors account for less than 1% of ovarian neoplasms (2). Most are

associated with virilization and, occasionally, with obesity, hypertension, and

glucose intolerance, reflecting corticosteroid secretion. Rare cases of estrogen

secretion and isosexual precocity have been reported.

Most of these tumors have benign or low-grade behavior, but about 20%,

most of which are initially larger than 8 cm in diameter, may metastasize.

Metastases are usually in the peritoneal cavity but can occur at distant sites,

including the liver. The primary treatment is surgical removal of the ovary. There

are no data regarding the effectiveness of radiation or chemotherapy for this

disease.

Sarcomas

Carcinosarcomas (malignant mixed mesodermal tumors) of the ovary are

uncommon. Clinical, morphologic, and molecular data suggest that they are

monoclonal and that both epithelial and mesenchymal components are derived

from a single uncommitted, totipotent cell which, after transformation, has

proliferated toward separate differentiation lineages. Carcinosarcomas are thought

to result from epithelial–mesenchymal transition (EMT) characterized by

epithelial plasticity with transdifferentiation toward a mesenchymal cell type.

Most tumors are heterologous, and 80% occur in postmenopausal women. The

signs and symptoms are similar to those of most ovarian malignancies. These

tumors are biologically aggressive, and most patients have evidence of metastases

at presentation. They are treated either with CP or ifosfamide and cisplatin, or

anthracycline-based combination regimens, but in general have a poor prognosis.

Small Cell Carcinoma, Hypercalcemic Type

This rare tumor occurs at an average age of 24 years (range 2 to 46 years)

(516). The tumors are typically unilateral. Approximately two-thirds of the

tumors are accompanied by paraendocrine hypercalcemia. This tumor

accounts for one-half of all of the cases of hypercalcemia associated with ovarian

tumors. About 50% of the tumors have spread beyond the ovaries when they are

diagnosed (516). Small cell carcinoma, hypercalcemic type is characterized by

germline or somatic mutations in SMARCA4. Germline and somatic SMARCA4

mutations characterize small cell carcinoma of the ovary, hypercalcemic type.

2653Similar mutations have been described in rhabdoid tumors and it has been

proposed that small cell ovarian cancers should be considered to be ovarian

rhabdoid tumors and treated as such (517,518).

Immunohistochemical stains are helpful to differentiate this tumor from a

lymphoma, leukemia, or sarcoma.

The management of these malignancies consists of surgery followed by

platinum-based chemotherapy, radiation therapy, or both. It has been

suggested that these patients may benefit from aggressive combination

chemotherapy regimens that have been used in rhabdoid tumors at other

sites. In addition to the primary treatment of the disease, control of the

hypercalcemia may require aggressive hydration, loop diuretics, and the use of

bisphosphonates or calcitonin. The prognosis tends to be poor, with most patients

dying within 2 years of diagnosis, in spite of treatment.

METASTATIC TUMORS

[14] About 5% to 6% of ovarian tumors are metastatic from other organs,

most frequently from the female genital tract, the breast, or the

gastrointestinal tract (519–533). The metastases may occur from direct

extension of another pelvic neoplasm, by hematogenous or lymphatic spread, or

by transcoelomic dissemination, with surface implantation of tumors that spread

in the peritoneal cavity.

Gynecologic

Nonovarian cancers of the genital tract can spread by direct extension or

metastasize to the ovaries. Tubal carcinoma involves the ovaries secondarily

in 13% of cases, usually by direct extension (2,4). Under some circumstances, it

is difficult to know whether the tumor originated in the tube or the ovary, when

both are involved. Cervical cancer spreads to the ovary only in rare cases (<1%),

and most of these are of an advanced clinical stage or are adenocarcinomas.

Adenocarcinoma of the endometrium can spread and implant directly onto the

surface of the ovaries in about 5% of cases, but two synchronous primary tumors

probably occur with greater frequency (532). In these cases, an endometrioid

carcinoma of the ovary is usually associated with the adenocarcinoma of the

endometrium.


FIGURE 39-25 Metastatic carcinoma in the ovary. Note the linear, single-cell pattern

found in this metastatic breast carcinoma.

Nongynecologic

The frequency of metastatic breast carcinoma to the ovaries varies according

to the method of determination, but the phenomenon is common (Fig. 39-25).

In the autopsy data of women who died of metastatic breast cancer, the

ovaries were involved in 24% of cases, and 80% of the involvement was

bilateral (519–524). Similarly, when ovaries are removed to palliate advanced

breast cancer, about 20% to 30% of the cases reveal ovarian involvement, 60% of

those bilaterally. The involvement of ovaries in early-stage breast cancer seems to

be considerably lower, but precise figures are not available. In almost all cases,

ovarian involvement is occult or a pelvic mass is discovered after other metastatic

disease becomes apparent.


FIGURE 39-26 Krukenberg tumor of the ovary metastatic from a gastric carcinoma.

Malignant cells have discrete vacuoles that push nuclei eccentrically, giving a signet-ring

appearance. Mucicarmine stain demonstrates the cytoplasmic vacuoles to be mucin.

Krukenberg Tumor

Krukenberg tumors are characterized by mucin-filled signet ring cells and

make up 30% to 40% of metastatic cancers to the ovaries (Fig. 39-26)

(525,526,534). The primary tumor is frequently located in the stomach and less

commonly in the colon, appendix (the so-called goblet cell carcinoid), breast, or

biliary tract. Rarely, the cervix or the bladder may be the primary site.

Krukenberg tumors can account for about 2% of ovarian cancers, and they are

usually bilateral. They are associated with a very poor prognosis and most

patients die of their disease within 1 year. In some cases, a primary tumor is never

found.

Other Gastrointestinal Tumors

2656In other cases of metastasis from the gastrointestinal tract to the ovary, the

tumor does not have the classic histologic appearance of a Krukenberg

tumor; most of these are from the colon and, less commonly, the

pancreatobiliary tract, appendix, and small intestine (Fig. 39-27). As many as

1% to 2% of women with intestinal carcinomas will develop metastases to

the ovaries during the course of their disease (521). Before exploration for an

adnexal tumor in a woman older than 40 years, a barium enema or colonoscopy is

recommended to exclude a primary gastrointestinal carcinoma with metastases to

the ovaries, if there are any gastrointestinal symptoms. Metastatic colon cancer

can mimic a mucinous cystadenocarcinoma of the ovary histologically, and the

histologic distinction between the two can be difficult (520,521,526–530).

Tumors that arise in the appendix may be associated with ovarian metastasis and

can be confused with primary ovarian mucinous cancer. Pseudomyxoma peritonei

almost always is associated with an appendiceal primary and the appendix should

always be resected at surgery and carefully examined pathologically (526,530).

Melanoma

Rare cases of malignant melanoma metastatic to the ovaries have been

reported (533). In these circumstances, the melanomas are usually widely

disseminated. Removal would be warranted for palliation of abdominal or pelvic

pain, bleeding, or torsion. Malignant melanoma can arise, rarely, in a mature

cystic teratoma (535).

Carcinoid Tumors

Metastatic carcinoid tumors represent fewer than 2% of metastatic lesions to

the ovaries (536). Only about 2% of patients with primary carcinoids have

evidence of ovarian metastasis, and only 40% of them have the carcinoid

syndrome at the discovery of the metastatic carcinoid. In perimenopausal and

postmenopausal women explored for an intestinal carcinoid, it is reasonable to

remove the ovaries to prevent subsequent ovarian metastasis. The discovery of an

ovarian carcinoid should prompt a careful search for a primary intestinal lesion

(537).


FIGURE 39-27 Metastatic colorectal carcinoma in the ovary often has areas of necrotic

debris (so-called dirty necrosis) adjacent to partial gland structures showing a cribriform

pattern.


FIGURE 37-28 Carcinoma of the fallopian tube. This is a high-grade serous carcinoma

that has invaded the lamina propria of the tubal mucosa. Most primary fallopian tube

carcinomas arise in the distal (fimbria) portion of the fallopian tube.

Lymphoma and Leukemia

Lymphomas and leukemia can involve the ovary. When they do, the

involvement is usually bilateral (538–540). About 5% of patients with Hodgkin

lymphoma will have lymphomatous involvement of the ovaries, but this

involvement typically occurs with advanced-stage disease. With Burkitt

lymphoma, ovarian involvement is very common. Other types of lymphoma

involve the ovaries less frequently, and leukemic infiltration of the ovaries is

uncommon (540). Sometimes the ovaries can be the only apparent sites of

involvement of the abdominal or pelvic viscera with a lymphoma; if this

circumstance is found, a careful surgical exploration may be necessary.

Intraoperatively, a hematologist–oncologist should be consulted to determine the

need for these procedures if frozen section of a solid ovarian mass reveals a

lymphoma. Most lymphomas no longer require extensive surgical staging; biopsy

of enlarged lymph nodes should be performed. In some cases of Hodgkin

2659lymphoma, a more extensive evaluation may be necessary. Treatment involves

that of the lymphoma or leukemia. Removal of a large ovarian mass may improve

patient comfort and facilitate a response to subsequent radiation or chemotherapy

Nhận xét