Adenocarcinoma of the uterine corpus and sarcomas of the uterus


Key points

  • 1.

    Adenocarcinoma of the endometrium is the most common gynecologic malignancy, with both rising incidence and mortality.

  • 2.

    Classically, endometrial cancer is divided into two histologic types: type I is endometrioid, and type II consists of uterine serous carcinoma, clear cell (CC), and other more aggressive histologic types, although the molecular classification is emerging as an important paradigm.

  • 3.

    The mainstay of treatment is surgery with removal of the uterus, cervix, fallopian tubes, and ovaries with incorporation of sentinel lymph node biopsy.

  • 4.

    Adjuvant therapy is sometimes given. Radiation, chemotherapy, or a combination of both is used depending on the clinical scenario.

  • 5.

    Some endometrial cancers (5%) result from inherited deoxyribonucleic (DNA) mismatch repair defects, known as Lynch syndrome. All patients with endometrial cancer should be evaluated for this possibility.

  • 6.

    Immunotherapy has been identified as an effective treatment strategy in patients with recurrent mismatch repair deficiency/microsatellite instability-high endometrial cancer.

  • 7.

    Uterine sarcomas are uncommon tumors. The most common tumors are leiomyosarcomas (LMSs; 59%) and endometrial stromal sarcomas (ESSs; 33%). Other sarcomas comprise less than 8% of the remaining.

  • 8.

    The standard treatment for any sarcoma is removal of the uterus, fallopian tubes, and ovaries.

  • 9.

    The prognosis of LMSs is poor; even at an early stage, adjuvant therapy has not been shown to affect survival.

  • 10.

    ESSs are often hormonally responsive.

Incidence

In the United States, cancer of the uterine corpus is the most common malignancy unique to women. It was estimated by the American Cancer Society that uterine cancer will develop in approximately 66,570 women in 2021 in the United States, making it the fourth most common cancer in women behind breast, lung, and colorectal cancer (Siegel 2021, #2597; ) ( Fig. 5.1 ). Importantly, however, when corrected for hysterectomy rates, uterine cancer becomes the second most commonly diagnosed cancer among women. The increased incidence of carcinoma of the endometrium has been apparent only during the past 3 decades. In reviewing the predicted incidence for the 1970s, the American Cancer Society noted a 1.5-fold increase in the number of patients with endometrial cancer; however, there was a decline in incidence during the late 1980s. More recently, deaths from uterine cancer have increased. In 1990, the American Cancer Society estimated 4000 deaths from this cancer, increasing to 10,470 in 2016, and most recently 12,940 in 2021 . Despite an overall improvement in cancer-related survival, endometrial cancer mortality continues to rise (Siegel, 2021, #2597).

Figure 5.1, Common new cancer cases and deaths in women for 2010 in the United States.

The increasing prevalence of overweight and obesity in women, especially in developed countries, may explain a component of the increased incidence of endometrial cancer. In the United States, approximately 57% of all endometrial cancers are attributed to obesity, with the risk of endometrial cancer increasing by more than 50% for each 5-unit increase in body mass index (BMI) ( ; ). Importantly, rate increases appear greatest in the non-endometrioid, type 2, endometrial cancers, including grade III endometrioid, serous, clear cell, and carcinosarcoma histologies, potentially reflecting the relevance of alternate factors independent of BMI ( ).

Epidemiology

Endometrial adenocarcinoma (AC) occurs during the reproductive and menopausal years. The mean age for patients with AC of the uterine corpus is 63 years; most patients are between the ages of 50 and 59 years ( Fig. 5.2 ). Approximately 5% of women will have AC before the age of 40 years, and 20% to 25% will be diagnosed before menopause. Bokhman suggested that there are two pathogenic types of endometrial cancer. The first type arises in women with obesity, hyperlipidemia, and signs of hyperestrogenism, such as anovulatory uterine bleeding, infertility, late onset of menopause, and hyperplasia of the stroma of the ovaries and endometrium. The second pathogenic type of disease arises in women who have none of these disease states or in whom the disease states are not clearly defined. Bokhman’s data suggest that patients with the first pathogenic type mainly have well differentiated or moderately differentiated tumors, superficial invasion of the myometrium, high sensitivity to progestins, and a favorable prognosis (85% 5-year survival rate in his material). The patients who fall into the second pathogenic group tend to have poorly differentiated tumors, deep myometrial invasion, a high frequency of metastatic disease in the lymph nodes, decreased sensitivity to progestin, and poor prognosis (58% 5-year survival rate).

Figure 5.2, Carcinoma of the corpus uteri; patients treated in 1999 to 2001. Age distribution by mode of staging.

Multiple risk factors for endometrial cancer have been identified, and MacMahon divides them into three categories:

  • Variants of normal anatomy or physiology

  • Frank abnormality or disease

  • Exposure to external carcinogens

Obesity, nulliparity, and late menopause are variants of normal anatomy or physiology classically associated with endometrial carcinoma. These three factors are evaluated in regard to the possible risk of developing endometrial cancer ( Table 5.1 ). If a patient is nulliparous and obese and reaches menopause at age 52 years or older, she appears to have a fivefold increase in the risk of endometrial cancer above that of a patient who does not satisfy these criteria ( Table 5.2 ).

TABLE 5.1
Endometrial Cancer Risk Factors
Risk Factors Risk
  • Obesity

  • 2.5–4.5×

  • Nulliparity

    • Compared with one child

    • Compared with five or more children

  • Late menopause

  • 2.4×

TABLE 5.2
Multiple Risk Factors
RISK
Nulliparous Parous
Top 15% in weight 5× more than Lower two thirds in weight
Menopause at 52 years Menopause before 49 years

The type of obesity in patients with endometrial cancer has been evaluated. In a study from the University of South Florida, it was noted that women with endometrial cancer had greater waist-to-hip circumference ratios, abdomen-to-thigh skin ratios, and suprailiac-to-thigh skin ratios than those of matched control women. As these ratios increased, the relative risk (RR) of endometrial cancer increased. The researchers concluded that upper-body fat localization is a significant risk factor for endometrial cancer. In a large multicenter case-control study of 403 endometrial cancer cases and 297 control cases, Swanson and associates confirmed and amplified these findings. Women whose weight exceeded 78 kg had a risk 2.3 times that of women weighing less than 58 kg. For women weighing more than 96 kg, the RR increased to 4.3. Upper-body obesity (waist-to-height ratio) was a risk factor independent of body weight. Patients in the highest quartile of both weight and waist-to-thigh circumference had a risk of 5.8 times. The amount of body fat has been associated with decreased circulating levels of both progesterone and sex hormone–binding proteins. There was a strong inverse association between sitting height and risk of endometrial cancer. This may be related to sex hormone–bound globulin (SHBG), which appears to be depressed in women with endometrial cancer. The level of SHBG is progressively depressed with increasing upper-body fat localization. With lower SHBG, there is a higher endogenous production of non–protein-bound estradiol. Because endometrial cancer is related to obesity, dietary habits appear to be important. Data suggest that the levels of estriol, total estrogens, and prolactin were lower and those of SHBG were higher in postmenopausal women who were vegetarians. In a case-control study, Levi and colleagues evaluated dietary factors in 274 patients with endometrial cancer and 572 control subjects from two areas in Switzerland and northern Italy. Extensive dietary history was obtained. Their data confirmed the relationship between obesity and endometrial cancer. In relation to diet, they noted an increased association with total energy intake. After correction for total energy intake, a risk was present with the frequency of consumption of most types of meats, eggs, beans, added fats, and sugar. Conversely, significant protection was noted with an elevated intake of most vegetables, fresh fruits, whole grain bread, and pasta. This reflected a low risk with increased intake of ascorbic acid and beta-carotene. Of dietary interest is that the intake of olive oil seemed beneficial in Switzerland but resembled other added fats in Italian women. It has been previously noted that the amount and type of dietary fat influence estrogen metabolism because estrogen reabsorption from the bowel seems to be increased by diets rich in beef or fats.

Diabetes mellitus and hypertension are frequently associated with endometrial cancer. Elwood and colleagues reported a RR of 2.8 associated with a history of diabetes after controlling for age, body weight, and socioeconomic status. High levels of insulin-like growth factor I, coupled with elevated estrogen levels, are thought to have neoplastic potential that accounts for the observed increased risk of endometrial cancer. High blood pressure is prevalent in older obese patients but does not appear to be a significant factor by itself, even though 25% of patients with endometrial cancer have hypertension or arteriosclerotic heart disease.

As extensively detailed in Chapter 4 , the relationship of unopposed estrogen and endometrial cancer is well documented. Fortunately, the addition of a progestin appears to be protective. The adequacy of progesterone is important in prevention of endometrial cancer. In a study from Sweden, at the end of 5 years, excess risk of endometrial cancer was 6.6, but with combined estrogen progestin (E + P), the RR was 1.6 for 11 to 15 days of progestin, 2.9 for 10 days of use, and 0.2 if continuous E + P was given. The Million Women study from the United Kingdom reported its findings of endometrial cancer and hormone replacement therapy (HRT). This study, which first reported on HRT and breast cancer, has been severely criticized mainly on methodologic factors. The study had an average follow-up of 3.4 years, during which 1320 incident endometrial cancers were diagnosed. At time of recruitment, 22% of HRT users (total number was 320,953 women) last used continuous combined therapy, 45% last used cyclic combined therapy with progestogen usually added for 10 to 14 days per month, 19% last used tibolone, and 4% used estrogen alone. Compared with nonusers, the RRs of endometrial cancer were 0.71, confidence interval (CI) 0.56 to 0.90, P = .005; 1.05, CI 0.91 to 1.22; 1.79, CI 1.43 to 2.25, P < .001; and 1.45, CI 1.02 to 2.06, P = .04, respectively.

Of note, the adverse effects of tibolone and estrogen only were greatest in the nonobese women, and the beneficial effects of combined HRT were greatest in obese women. Although the risk of unopposed estrogen is present, women taking estrogen who develop endometrial cancer appear to have favorable prognostic factors. Several, but not all, studies suggest that risk factors such as multiparity and obesity are lower in the estrogen users. Stage of disease and histologic grade appear to be lower in estrogen users. With correction for stage and grade, estrogen users still have less myometrial invasion than non-estrogen users do. The poor prognostic subtypes, such as clear cell carcinoma, appear less frequently in estrogen users. As a result, survival rates with estrogen-related endometrial cancer are much better than those of non–estrogen-related cancers.

Data indicate that the use of combination oral contraceptives decreases the risk for development of endometrial cancer. The Centers for Disease Control and Prevention evaluated endometrial cancer cases of all women aged 20 to 54 years from eight population-based cancer registries and compared them with control patients selected at random from the same centers. A comparison of the first 187 cases with 1320 control cases showed that women who used oral contraceptives at some time had a 50% reduction in the risk of developing endometrial cancer compared with women who had never used oral contraceptives. This protection occurred in women who used oral contraceptives for at least 12 months, and protection continued for at least 10 years after oral contraceptive use. Protection was most notable for nulliparous women. These investigators estimated that about 2000 cases of endometrial cancer are prevented each year in the United States by past or current use of oral contraceptives. Of interest, cigarette smoking appears to decrease the risk for developing endometrial cancer, although given the multiple cancers and other diseases associated with smoking this would not be advocated as a risk-reducing measure. In a population-based case-control study of women aged 40 to 60 years, Lawrence and associates found a significant decline in RR of endometrial carcinoma with increased smoking ( P > .05).

Incidence and survival are higher in White women compared with Black women. Reasons for these differences are unexplained. An analysis of the Gynecologic Oncology Group (GOG) database evaluated this factor in 600 White and 91 Black women with clinical stage I or stage II endometrial cancer. A larger number of the African American women were diagnosed after age 70 years, and they had a higher proportion of serous and clear cell histologies; Black women also had more advanced disease, higher grade, vascular space involvement, depth of invasion, and lymph node metastases than the White women did. The survival rate (5-year) was 77% for White women and 60% for Black women. Survival difference remained even in high-risk groups such as grade III tumors (59% vs. 37%, respectively). Clarke and colleagues suggest that hysterectomy-corrected incidence rates of uterine corpus cancer were similar among non-Hispanic White and Black women and lower among Hispanics and Asian/Pacific Islanders ( ). Furthermore, non-endometrioid carcinoma and sarcoma rates were highest in non-Hispanic Black women, with the lowest survival rates in this population irrespective of stage at diagnosis or histologic subtype, highlighting the importance of continued investigation into racial and ethnic differences in oncologic outcome ( ). In non-Hispanic Black patients, the overall 5-year survival rate was 63.2%, compared to 86.1% in non-Hispanic White patients, and 81.4% in Hispanic patients ( Fig. 5.3 ). Data emerging from this study additionally suggest that the increase in non-endometrioid cancer cases, combined with stable rates of endometrioid cancers argue against the obesity epidemic as the sole driver of the rising uterine cancer incidence ( ).

Figure 5.3, Trends in age-adjusted incidence rates of microscopically confirmed uterine corpus cancer by race and ethnicity: (A, B) overall and by (C, D) endometrioid and (E, F) nonendometrioid subtypes, (A, C, E)

Tamoxifen is used to prevent or treat breast cancer. Tamoxifen was first introduced in clinical trials in the early 1970s and was approved in 1978 by the US Food and Drug Administration (FDA) for treatment of advanced breast carcinoma in postmenopausal woman. Tamoxifen, although labeled an antiestrogen, is known to have estrogenic properties and truly is a weak estrogen. Women receiving tamoxifen also appear to have some protection from osteoporosis and heart disease (decreased lactate dehydrogenase and cholesterol), similar to women receiving estrogen replacement therapy. Extensive experience with this drug has been reported. It is estimated that more than 4 million women in the United States have taken tamoxifen for almost 8 million women-years of use. One of its major benefits is that in women taking tamoxifen, there has been a substantial decrease in the incidence of a second cancer in the opposite breast compared with similar women who were taking a placebo.

The Early Breast Cancer Trialists Collaborative Group (EBCTCG) has produced an important meta-analysis of 194 randomized trials of adjuvant chemotherapy or endocrine therapy with at least 15 years of follow-up. The analysis evaluated the effects of adjuvant tamoxifen on breast cancer recurrence and survival. It was shown that 5 years of tamoxifen therapy, compared with no adjuvant therapy, reduced the 15-year probability of breast cancer recurrence (from 45% to 33%) and breast cancer mortality (from 35% to 26%). In addition, tamoxifen has been shown to provide a preventive benefit in women at risk for developing breast cancer.

There has been a considerable amount of discussion in the literature concerning the association of tamoxifen with endometrial cancer. At least three studies (Fisher and colleagues; Powels and colleagues; Veronesi and colleagues) evaluating the prophylactic use of tamoxifen in women without breast cancer have reported an association between tamoxifen use and endometrial cancer. In addition, several cases of endometrial cancer have been described in women receiving tamoxifen. In a prospective, randomized study of the National Surgical Adjuvant Breast and Bowel Project (NSABP), 2843 patients with node-negative estrogen receptor–positive invasive breast cancer were randomly assigned to receive a placebo or 20 mg/day of tamoxifen. An additional 1220 tamoxifen-treated patients were registered and given the drug. The average time in the study was 8 years for the randomly assigned patients and 5 years for the registered patients. Of the 1419 patients randomly assigned to tamoxifen, 15 developed uterine cancer, of which two were sarcomas. One patient randomly assigned to receive tamoxifen did not take the drug and developed endometrial cancer 78 months after randomization. In the placebo group, two developed endometrial cancer; however, both were receiving tamoxifen at the time of their uterine malignant disease. One patient had a breast recurrence and was prescribed tamoxifen, and the other was given tamoxifen after colon cancer. Two of the patients with endometrial cancer had been taking tamoxifen for only 5 and 8 months before their diagnosis of uterine disease was made. Five patients in the tamoxifen group developed endometrial cancer after the drug had been discontinued for 7 to 73 months. In the registered patients who received tamoxifen, eight uterine tumors (seven endometrial) were subsequently diagnosed. Three of these patients had been taking tamoxifen for less than 1 year (2 months, 2 months, and 9 months). The authors determined the average annual hazards ratio (HR) of endometrial cancer per 1000 women in their population of patients. This was 0.2 per 1000 in the placebo group and 1.6 per 1000 for the randomized tamoxifen-treated patients. In the registered patients receiving tamoxifen, the average annual hazard rate was 1.4 per 1000, similar to that of the randomized tamoxifen-treated group. The hazard rate of endometrial cancer in the placebo group was low compared with the Surveillance, Epidemiology, and End Results (SEER) data and with previous NSABP randomized tamoxifen–placebo studies; these data suggest that the average annual hazard rate is 0.7 per 1000.

These data, based on a limited number of patients with endometrial cancer while receiving tamoxifen, suggest that there may be an RR of 2.3 for development of endometrial cancer while receiving tamoxifen. This does not take into account the well-known fact that women who develop breast cancer are at an increased risk for development of endometrial cancer irrespective of subsequent treatment. A RR of 1.72 to more than 3 has been reported. The risks and benefits of the prevention of recurrences and new breast cancer in comparison to new endometrial cancers were evaluated in the NSABP study. The benefits suggest that 121 fewer breast-related events per 1000 women treated with tamoxifen were seen compared with 6.3 endometrial cancers per 1000 women. Therefore, the benefit from tamoxifen is apparent.

It was initially suggested that the rate of endometrial cancers associated with tamoxifen use might be equal to that associated with unopposed estrogen replacement therapy. Because tamoxifen is a weak estrogen, similar characteristics of endometrial cancer were also implied (i.e., well-differentiated superficially invasive cancers). Barakat and associates reviewed five studies, including the study by Magriples, the NSABP, their own data from Memorial Sloan-Kettering Hospital, and two studies from overseas. A total of 103 patients were evaluated in regard to histologic features, grade of tumor, International Federation of Gynecology and Obstetrics (FIGO) staging, and deaths from uterine cancer; an increase was not found in poor prognostic histologic findings, tumor differentiation, or stage compared with what would be expected in a similar group of non–tamoxifen-treated patients with uterine cancer. Jordan, in an evaluation of the SEER data and of tamoxifen-associated endometrial cancer in the literature, reported similar findings.

It is suggested that all women, irrespective of whether they are taking tamoxifen, should have yearly gynecologic examinations. The endometrium should be evaluated, via biopsy, if the patient is symptomatic. We concur with the American College of Obstetricians and Gynecologists (ACOG) committee opinion that recommends against endometrial sampling or ultrasound evaluation of the endometrium just because an individual is taking tamoxifen. This possible concern of tamoxifen and endometrial cancer may lessen in the near future because the aromatase inhibitors (AIs) may appear to be better than tamoxifen in the prevention of recurrent or contralateral breast cancer. Several clinical trials have demonstrated the comparable if not greater efficacy of AIs compared with tamoxifen. Although tamoxifen remains an option for adjuvant therapy for postmenopausal women, AIs are thought to be more effective in preventing breast cancer recurrence in the first 2 years after surgery. AIs reduce estrogen levels in postmenopausal women by inhibiting or inactivating aromatase, the enzyme that synthesizes estrogens from circulating androgens. AIs should be avoided in premenopausal women, including those who have experienced chemotherapy-induced amenorrhea. Whereas tamoxifen is a partial agonist, AIs are not agonists and are not associated with estrogenic-related thromboembolic events and uterine cancers. The activity of third-generation agents anastrozole, letrozole, and exemestane is generally considered comparable. Although AIs are associated with a significant risk of osteoporosis, they do not increase the risk of gynecologic problems. In one large adjuvant therapy trial (the Anastrozole, Tamoxifen Alone or in Combination Trial [ATAC]), anastrozole was associated with fewer cerebrovascular events (2.0% vs. 2.8%), endometrial cancers (0.2% vs. 0.8%), thromboembolism (3% vs. 4%), and vaginal bleeding (5% vs. 10%) compared with tamoxifen. The role of AIs as prophylaxis has been studied, showing a reduction in new breast cancers relative to placebo, although there are no AIs approved by the US FDA for this indication.

Although the majority of endometrial cancer cases are sporadic, hereditary endometrial cancer has been identified in association with hereditary nonpolyposis colon cancer (HNPCC), also known as Lynch II syndrome. This is an autosomal-dominant inherited cancer that involves a germline mutation in one of the genes in the DNA mismatch repair (MMR) gene family, which includes MSH2, MLH1, PMS2, and MSH6 . Fortunately, HNPCC accounts for only 1% to 5% of all colorectal cancers but it is associated with a 39% to 54% lifetime risk of developing colon cancer. There is a lifetime risk of 30% to 61% of developing endometrial cancer in those with HNPCC. There is also an increased risk of ovarian cancers and other non-gynecologic cancers. In a study by Lu and associates, they noted that about half the time, the endometrial or ovarian cancer occurs prior to the colon cancer. In both instances, the age at diagnosis was in the early 40s. There was a median of 11 years between the gynecologic cancer and colon cancer diagnosis. About 14% of the time, the gynecologic and colon cancers were diagnosed simultaneously. Several “red flags” should prompt an evaluation for HNPCC. These include any individual with a personal or family history of colon cancer at an early age of onset (usually before age 50 years) or endometrial cancer at an early age of onset (premenopausal or before age 50 years) and two or more HNPCC-related cancers in an individual or family. Assessing a patient’s risk for hereditary cancer is an important process, beginning with screening for the “red flags” of hereditary colon (and endometrial) cancer. Individuals with any of the “red flags” should enter into a discussion about genetic testing to determine if this is appropriate for them. Medical management strategies can be tailored depending on the genetic testing results and may include increased surveillance, chemoprevention, and prophylactic surgery. The Cancer Study Consortium suggests colonoscopy every 1 to 3 years beginning at age 25 years in individuals with this hereditary disorder. The data suggest that if surveillance is done, survival rates are improved. Women should be offered surveillance with ultrasound and endometrial sampling from age 25 to 35 years, although there are no data to suggest this will improve survival if endometrial cancer is diagnosed by these means. As reported by Schmeler and colleagues, risk-reducing surgery consisting of removal of the uterus, fallopian tubes, and bilateral ovaries has been shown to decrease the risk of uterine and ovarian cancer in these high-risk patients. Incorporation of MMR immunohistochemical (IHC) testing of all endometrial cancer specimens is now advocated to both inform therapeutic strategies, and to simultaneously assess for the possibility of underlying Lynch syndrome versus somatic or epigenetic alterations .

Diagnosis

Routine screening for uterine AC and its precursors is not recommended. Women receiving HRT (estrogen and progesterone) do not need endometrial biopsy before institution of therapy or during replacement therapy unless abnormal bleeding occurs. Monthly withdrawal bleeding after progestin is not considered abnormal bleeding. However, breakthrough bleeding should be evaluated. The use of continuous estrogen alone increases the risk of AC. Estrogen plus progesterone appears to decrease the risk of AC and therefore is the preferred treatment. In asymptomatic high-risk patients, periodic evaluation may be advisable. All postmenopausal women with uterine bleeding must be evaluated for endometrial cancer, although only 20% of these patients will have a malignant genital neoplasm. As the patient’s age increases after menopause, there is a progressively increasing probability that her uterine bleeding is caused by endometrial cancer. Feldman and associates found that age was the greatest independent risk factor associated with endometrial cancer or complex hyperplasia. In women aged 70 years or older, the odds ratio was 9.1. If complex hyperplasia was present, the odds ratio increased to 16. When a woman was older than 70 years, her chance of having cancer when vaginal bleeding was present was about 50%. If she was also nulliparous and had diabetes, the risk was 87%. A perimenopausal patient who may have abnormal uterine bleeding indicative of endometrial cancer is frequently not evaluated because the patient or her physician interprets her new bleeding pattern as resulting from menopause. During this time in a woman’s life, the menstrual periods should become progressively lighter and farther apart. Any other bleeding pattern should be evaluated with carcinoma of the endometrium in mind. A high index of suspicion must be maintained if the diagnosis of endometrial cancer is to be made in a young patient. Prolonged and heavy menstrual periods and intermenstrual spotting may indicate cancer, and endometrial sampling is advised. Most young patients who develop endometrial cancer are obese, often with anovulatory menstrual cycles.

Historically, fractional dilation and curettage (D&C) has been the definitive diagnostic procedure used in ruling out endometrial cancer. Today, most advocate the routine use of the endometrial biopsy as an office procedure to make a definitive diagnosis and spare the patient hospitalization and need for anesthetic. Several studies have indicated that the accuracy of the endometrial biopsy in detecting endometrial cancer is approximately 90%. Cytologic detection of endometrial cancer by routine cervical Papanicolaou (Pap) smear has generally been poor in comparison with the efficacy of the Pap smear in diagnosing early cervical neoplasia. Several studies in the literature indicate that only one-third to half of the patients with AC of the endometrium have abnormal Pap smear results on routine cervical screening. The main reason for the poor detection with the cervical Pap smear is that cells are not removed directly from the lesion. When a cytologic preparation is obtained directly from the endometrial cavity, malignant cells are present in higher numbers than those found if routine cervical or vaginal smears are obtained. Techniques that obtain only a cytologic preparation are generally inadequate if they are used alone.

Several commercial apparatuses are available for sampling the endometrial cavity on an outpatient basis. If diagnosis of endometrial cancer can be made on an outpatient basis, the patient can avoid hospitalization and a minor surgical procedure. Devices that remove tissue for histologic evaluation have generally been good if tissue is obtained from the endometrial cavity. Stovall and colleagues used the Pipelle instrument to evaluate 40 patients known to have endometrial cancer. Ninety percent of the women were postmenopausal. Only in one patient was cancer not identified with the Pipelle. This patient had a prior D&C that revealed a grade I lesion. The Pipelle diagnosis was atypical adenomatous hyperplasia, and the hysterectomy specimen revealed a focus of AC in situ. The pathologist noted that the obtainable tissue was acceptable for analysis in 100% of patients. Discomfort was recorded as mild in 80%, and only two patients (5%) reported severe pain. Goldchmit and coworkers reported similar accuracy with the Pipelle in 176 consecutive patients undergoing D&C. Whereas endometrial biopsy and D&C appear to be equivalent in terms of diagnosing cancer, the accuracy of endometrial biopsy appears to be inferior to D&C in predicting final post-hysterectomy tumor grade. In a recent study by Leitao and colleagues, 18% of endometrial biopsy specimens were upgraded on final hysterectomy specimen, but only 9% of D&C specimens were upgraded. In symptomatic patients in whom inadequate tissue (or no tissue at all) is obtained for pathologic evaluation or in patients with persistent postmenopausal bleeding and non-diagnostic endometrial biopsy, a D&C must be considered.

Hysteroscopy has been suggested as an adjuvant in making the diagnosis of endometrial cancer and in establishing the extent of disease. Hysteroscopy has been used frequently in the evaluation of patients with abnormal uterine bleeding and has the advantages of allowing the physician to see the pathologic lesion and direct biopsy, identify other competing diagnoses (fibroids, polyps), and perform the procedure on an outpatient basis. Clark and colleagues analyzed data from 65 primary studies on the use of hysteroscopy to diagnose endometrial cancer and endometrial disease (cancer, hyperplasia, or both), including more than 26,000 women. All of the patients had abnormal premenopausal or postmenopausal uterine bleeding. Using endometrial histologic findings as a reference, a positive hysteroscopy result was associated with a 72% probability of endometrial cancer, but a negative result reduced this probability to 0.6%. The corresponding probabilities for endometrial disease were 55% with a positive result and 3% with a negative result. The accuracy of hysteroscopy tended to be higher among postmenopausal women and in an outpatient setting. They concluded that hysteroscopy is highly accurate and thereby clinically useful in diagnosing endometrial cancer in women with abnormal uterine bleeding and is moderately useful in diagnosing endometrial disease. Because many patients with endometrial cancer can be diagnosed with outpatient office biopsy, this remains the preferred first diagnostic step. If the biopsy result is negative and further evaluation is needed, we proceed to hysteroscopy with endometrial curettage. With its use, surgeons can direct biopsies of focal lesions that might be missed by D&C. Hysteroscopy can also be used to evaluate the endocervical canal.

Ultrasonography has been suggested as a diagnostic tool in evaluating women with irregular bleeding, particularly postmenopausal patients ( Fig. 5.4 ). The endometrial stripe as seen with transvaginal ultrasonography appears to be indicative of endometrial thickness. Several studies suggest that if a thin endometrial stripe is present, a histologic diagnosis is not necessary because atrophic endometrium would be present. Granberg and associates evaluated 205 women with postmenopausal bleeding, 30 postmenopausal asymptomatic women, and 30 postmenopausal patients with known endometrial cancer. In the two groups of 60 patients, the endometrial thickness was 3.2 (mean) versus 17.7, respectively. In the group of 205 women, 18 were found to have endometrial cancer. No cancers were present in the endometrium that had an endometrial thickness of 8 mm or less. There was considerable overlap of endometrial thickness by all histologic groups. The authors noted that if a cutoff of 5 mm was used, no false-negative findings were present. With this measurement, the positive predictive value was 87%, with specificity of 96% and sensitivity of 100% for identifying endometrial abnormalities. It has been suggested that if ultrasonography could save a large number of endometrial biopsies, there would be a large cost savings with less discomfort to the patient. As previously noted, significant pain with the newer disposable endometrial biopsy techniques affects only a small number of patients, and a certain number of patients, because of considerable endometrial thickness, will require endometrial sampling anyway. Clark and colleagues investigated the cost-effectiveness of initial diagnostic strategies for postmenopausal bleeding. A decision analytic model was constructed to reflect current service provision, which evaluated 12 diagnostic strategies using endometrial biopsy, ultrasonography (4- and 5-mm endometrial thickness cutoff), and hysteroscopy. Diagnostic probability estimates were derived from systematic quantitative reviews, clinical outcomes from published literature, and cost estimates from local and National Health Service sources. The main outcome measure was the cost per additional life year gained (£/LYG). Compared with carrying out no initial investigation, a strategy based on initial diagnosis with ultrasonography using a 5-mm cutoff was the least expensive (£11,470/LYG). Initial investigation with endometrial biopsy or ultrasonography using a 4-mm cutoff was comparably cost-effective (<£30,000/LYG vs. ultrasonography with a 5-mm cut off). The strategies involving initial evaluation with test combinations or hysteroscopy alone were not cost-effective. They concluded that women presenting for the first time with postmenopausal bleeding should undergo initial evaluation with ultrasonography or endometrial biopsy.

Figure 5.4, A, Ultrasound image of the uterus showing the “triple line” indicating the thickness of the endometrium. B, Ultrasound of the uterus showing a “thickened endometrium” of more than 10 mm. C, Saline instillation of the endometrial cavity notes a well-defined submucous fibroid and not thickened endometrium.

Unfortunately, endometrial cancer has been identified when the endometrial thickness is less than 5 mm. Although studies may evaluate several hundred patients, most do not have many cancer patients included. Wang and associates reviewed the ultrasounds of 52 women who were diagnosed with serous uterine cancer, clear cell and other high-grade carcinomas. Of the 52, 34 (65%) had a thickened endometrium measuring 5 mm or more; in nine (17%) the endometrium was less than 5 mm, and in an additional nine women (17%) the endometrium was indistinct. In the women with a non-thickened endometrium, other ultrasound abnormalities were noted: intracavitary fluid or lesion, myometrial mass, enlarged uterus, or adnexal mass. With the increasing incidence of type 2 endometrial cancers, the use of ultrasound as a triage mechanism may become less reliable. Multiple factors can affect endometrial thickness. These include estrogen use, estrogen + progestin use, BMI, diabetes, poor histotype, race, and postmenopausal status. The Committee on Gynecologic Practice of the ACOG issued an opinion on the role of transvaginal ultrasonography in the evaluation of postmenopausal bleeding. They concluded that women with postmenopausal bleeding may be assessed initially with either endometrial biopsy or transvaginal ultrasonography. This initial evaluation does not require performance of both tests. Transvaginal ultrasonography can be useful in the triage of patients in whom endometrial sampling was performed but tissue was insufficient for diagnosis. When transvaginal ultrasonography is performed for patients with postmenopausal bleeding and an endometrial thickness of less than or equal to 4 mm is found, endometrial sampling is not required. Meaningful assessment of the endometrium by ultrasonography is not possible in all patients. In such cases, alternative assessment should be completed. When bleeding persists despite negative initial evaluations, additional assessment is required, especially to rule out a type 2 carcinoma.

The reliability of determining endometrial thickness in postmenopausal patients does not appear to be applicable to women taking tamoxifen. In all studies, the endometrium in a tamoxifen-treated patient is considerably thicker than in a non–tamoxifen-treated patient. Histologic evaluation revealed an atrophic endometrium in a large number of these tamoxifen-treated patients. Lahti and colleagues evaluated 103 asymptomatic postmenopausal patients (51 receiving tamoxifen and 52 control participants) with ultrasonography, hysteroscopy, and endometrial histologic examination. In the tamoxifen group, 84% had an endometrial thickness on ultrasonography of 5 mm or more versus 19% in the non-tamoxifen group (51% vs. 8% >10 mm, respectively). Hysteroscopy findings noted that 28% of uterine mucosa was atrophic versus 87% in the non-tamoxifen control group. Histopathologic examination noted atrophic endometrium in 60% of tamoxifen-treated patients versus 79% of control subjects. The biggest difference between the two groups was the finding of polyps in 18% of the tamoxifen group versus 0% of the control group; this appears to be a frequent finding in the tamoxifen-treated patient. So-called mega-polyps measuring up to 12 cm have been described. Other uterine diseases have been attributed to tamoxifen, including increased uterine volume, lower impedance to blood flow in uterine arteries, endometriosis, focal peri-glandular condensation of stromal cells, and epithelial metaplasia. Data now suggest that the markedly thickened endometrium (≤40 mm) in patients receiving tamoxifen is not thickened endometrium but proximal myometrium.

Ultrasonography has also been evaluated as a means for determining depth of myometrial invasion. Gordon and associates studied 15 known patients with endometrial cancer by ultrasonography and magnetic resonance imaging (MRI). By use of criteria of greater than 50% myometrial wall involvement as deep invasion and less than 50% as superficial invasion, ultrasonography was judged to be more accurate than MRI in five studies; MRI was better in three, both were equally accurate in four, and neither was accurate in three. It has been suggested by some that ultrasonography can accurately predict myometrial invasion in about 75% of cases. Although knowing the depth of invasion preoperatively would be important information to the clinician, the data from studies as noted before would currently appear to be too premature or too costly to use routinely and use may be further limited for type 2 carcinomas as prior studies have excluded or only had limited numbers included in the cited studies. Evaluation of depth of myometrial invasion intraoperatively with gross examination or frozen section is preferred over preoperative radiographic assessment. An exception to the above exists in patients who desire and are appropriate candidates (type 1 carcinomas) for fertility preserving management, where MRI can be obtained to inform myometrial invasion or cervical involvement. In a prospective study of 101 patients with histologically confirmed endometrial cancer, MRI had a sensitivity and specificity of 89% and 100%, respectively, in determining myometrial invasion ( ).

Pathology

Careful evaluation of the uterus by the pathologist is essential for proper diagnosis and treatment of corpus cancer ( Fig. 5.5 ). Gross inspection of a bivalved uterus at the time of hysterectomy can offer an impression of the size of the lesion, its location (involvement of fundus, lower uterine segment, or cervix), and the depth of tumor penetration into the myometrium (depth of invasion). A clinically enlarged uterus may be caused by increasing tumor volume, but this should not be the only gauge for significant local disease. Obviously, many patients can have enlarged uteri because of factors other than AC. Carcinoma of the endometrium may start as a focal discrete lesion, as in an endometrial polyp. It may also be diffuse in several different areas, in some situations involving the entire endometrial surface. As the tumor grows, it can become larger or spread within the endometrium or myometrium. Endometrial cancer may disseminate to regional lymph nodes, by embolization or direct extension into the pelvis or vagina, or hematogenously to distant organs ( Fig. 5.6 ). The risk of spread is related to several factors, including depth of invasion into the myometrium, tumor grade, and histologic type. Further adding to the complexity, is the observation that type 2 carcinomas may not have grossly visible tumor but can be deeply infiltrative microscopically.

Figure 5.5, Pathologic evaluation of endometrial cancer.

Figure 5.6, Spread pattern of endometrial cancer with particular emphasis on potential lymph node spread. Pelvic and periaortic nodes are at risk, even in stage I disease.

Endometrioid AC, the most common histologic type, is usually preceded by a predisposing lesion, atypical endometrial hyperplasia ( Table 5.3 ). Only hyperplasia with cellular atypia is considered a true “precancerous” precursor of AC of the endometrium. For most patients with endometrioid-type tumors, particularly grade I or II lesions, and hyperplasia, hyperestrogenism is the etiologic basis. Pathologically, endometrial cancer is characterized by the presence of glands in an abnormal relationship to each other, with the hallmark of little, if any, intervening stroma between the glands. There can be variations in the size of the glands, and infolding is common. The cells are usually enlarged, as are the nuclei, along with nuclear chromatin clumping and nucleolar enlargement. Mitosis may be frequent. Differentiation of AC (grades I, II, or III) is important prognostically and is incorporated into FIGO surgical staging ( Fig. 5.7 ). Most studies suggest that 60% to 65% of all endometrial cancers are of the endometrioid subtype.

TABLE 5.3
Endometrial Carcinoma Subtypes
From Creasman WT, Odicino F, Maisonneuve P, et al: Carcinoma of the corpus uteri, Int J Gynecol Obstet 83(Suppl 1):79–118, 2003. No abstract available.
Type Number (%)
Endometrioid 6231 (84)
Adenosquamous 317 (4.2)
Mucinous 74 (0.9)
Uterine papillary serous 335 (4.5)
Clear cell 185 (2.5)
Squamous cell 28 (0.04)
Other 285 (3.8)

Figure 5.7, Histologic patterns of differentiation in endometrial carcinoma. A, Well-differentiated (G1) . B, Moderately differentiated (G2). C, Poorly differentiated (G3).

For almost a century, it has been recognized that a squamous component may be associated with an AC of the endometrium. This occurs in about 25% of patients. Historically, patients with a squamous component were further stratified according to whether the squamous component appeared benign (designated adenoacanthoma [AA]) or malignant (designated adenosquamous carcinoma [AS]). It was suggested that AA indicated a good prognosis and those with AS had a poor survival. Today, this distinction has been questioned concerning its prognostic importance. Zaino and coworkers, in reporting data from the GOG, suggest that the notation of squamous component irrespective of differentiation does not affect survival. Patients with clinical stage I and stage II cancers were evaluated, and 456 with typical AC and 175 with squamous differentiation (AC + SQ) were identified. The latter were subdivided into 99 with AA and 69 with AS. Multiple known prognostic factors were compared with differentiation of glandular and squamous component of the tumor. Age, depth of myometrial invasion, architecture, nuclear grade, and combined grade were similar for AC and AC + SQ, although patients with AA were better differentiated than those with AS and had less myometrial invasion. Both glandular and squamous differentiation correlated with frequency of pelvic and paraaortic node metastasis. Nodal metastasis, when it was stratified for grade and depth of invasion, was similar in AC and AC + SQ patients. The differentiation of squamous component is closely correlated with the differentiation of the glandular element, and the glandular element is a better predictor of outcome. It would therefore appear that the previous designation of AA and AS has no added predictive property than differentiation of glandular component and probably should be dropped as a diagnostic term. The authors suggest the term squamous differentiation instead, with differentiation of the glandular component noted as the important prognostic factor.

Secretory AC ( Fig. 5.8 ) is an uncommon type of endometrial cancer. It usually represents well-differentiated carcinoma with progestational changes. It is difficult to differentiate from secretory endometrium. Survival is good and comparable to that associated with the pure AC. Although it is an interesting histologic variant, the separation of the entity as it relates to treatment and survival is probably not warranted.

Figure 5.8, High-power view of well-differentiated secretory carcinoma invading the inner third of the myometrium.

Increasing emphasis has been placed on the importance of uterine serous carcinoma (USC). This subtype represents approximately 5% to 10% of all ACs but is highly aggressive, representing up to 40% of endometrial cancer-related deaths. Unlike the more common variants, USC is not associated with hyperestrogenism and frequently develops in the setting of atrophic endometrium. It is more commonly seen in older and non-White patients. Hendrickson, in the early 1980s, noted that in more than 250 endometrial cancers, only 10% had histologic features of USC, but these accounted for 50% of all treatment failures. The histopathologic appearance resembles a high-grade serous carcinoma of the ovary that has a propensity for vascular or lymphatic vascular space involvement (LVSI), as well as extrauterine spread, which can occur in the absence of myometrial invasion. Well-formed papillae are lined by neoplastic cells with grade III cytologic features ( Fig. 5.9 ). Differentiation between papillary architecture and syncytial metaplasia with benign endometrial alterations must be made because the papillary architecture alone does not designate USC. The uterus may appear grossly normal, without a visible lesion, but can have extensive myometrial invasion. Most USC are aneuploid and have a high S-phase and can be pure or admixed with other histologic types (endometrioid, clear cell, carcinosarcoma). USC commonly arise in endometrial polyps, with varying degrees of myometrial invasions. Current models of pathogenesis hypothesize that USC is a progressive lesion, arising from endometrial intraepithelial carcinoma (EIC), a finding supported by molecular assessments of paired EIN and USC tissue samples ( ; ).

Figure 5.9, Serous adenocarcinoma. The similarity to ovarian carcinoma is apparent.

Clear cell carcinomas ( Fig. 5.10 ) are also infrequent, accounting for 1% to 6% of all endometrial cancer, with distinct histologic criteria. Clear cell tumors are characterized by large polyhedral epithelial cells that may be admixed with typical non–clear cell ACs. Some authorities accept the mesonephritic-type hobnail cells as part of this pattern, but others believe that this histologic type should be excluded from the clear cell category. Silverberg and DeGiorgi and Kurman and Scully suggested a worse prognosis for clear cell AC than for pure AC. This was confirmed in studies by Christopherson and coworkers. Even in stage I disease, only 44% of patients with clear cell carcinomas survive 5 years. Neither the FIGO classification nor nuclear grade correlates with survival. Photopulos and associates, in a review of their material, noted that patients with this entity were older and tended to have a worse prognosis. They did note that patients with stage I clear cell carcinomas had a 5-year survival rate similar to that of patients with stage I pure AC of the endometrium.

Figure 5.10, Clear cell carcinoma of the endometrium. Clear cell component is quite evident.

Tumor grade

In addition to histologic type, pathologists assign a measure of tumor differentiation, known as grade, to endometrial cancers. Grade I lesions are well differentiated, frequently associated with estrogen excess, closely resemble hyperplastic endometrium, and are generally associated with a favorable prognosis. Grade III lesions are poorly differentiated, do not resemble normal endometrium, and frequently have a poorer prognosis. Grade II tumors are moderately differentiated and have an intermediate prognosis. Both architectural criteria and nuclear grade are used to classify. Architectural grade is related to the proportion of solid tumor growth, with grade I having an AC with less than 5% of the tumor exhibiting solid sheets, grade II having 6% to 50% of the neoplasm arranged in solid sheets of neoplastic cells, and grade III having greater than 50% of the neoplastic cells in solid masses. Regions of squamous differentiation are excluded from this assessment. The FIGO rules for grading state that notable nuclear atypia, inappropriate for architectural grade, raises the grade of a grade I or grade II tumor by one. By convention, serous and clear cell histologies are considered grade III or high-grade tumors.

Prognostic factors

After hysterectomy and lymph node assessment, clinical-pathologic characteristics are commonly used to predict risk of recurrence and to optimize therapy. Multiple factors have been identified for endometrial carcinoma that have prognostic value ( Table 5.4 ). Essentially all reports in the literature agree stage (extent of disease spread), tumor grade, and depth of invasion are important prognostic considerations. Before 1988, endometrial cancer was clinically staged with stage assignments based on uterine size and clinical extent of disease. Because of the considerable discrepancy between the clinical extent of disease spread and pathologic spread noted after surgical staging, FIGO adopted a surgical-pathologic staging classification in 1988. In 2009, FIGO updated the staging system ( Table 5.5 ). FIGO staging classification attempts to categorize patients into prognostic groups based on extent of disease and tumor grade.

TABLE 5.4
Prognostic Factors in Endometrial Adenocarcinoma
  • Histologic type (pathology)

  • Histologic differentiation

  • Stage of disease

  • Myometrial invasion

  • Peritoneal cytology

  • Lymph node metastasis

  • Adnexal metastasis

TABLE 5.5
2009 International Federation of Gynecology and Obstetrics Staging System for Carcinoma of the Endometrium
Stage I a Tumor Contained to the Corpus Uteri
Ia No or less than half myometrial invasion
Ib Invasion equal to or more than half of the myometrium
Stage II Tumor invades the cervical stroma but does not extend beyond the uterus b
Stage III a Local and/or regional spread of tumor c
IIIa Tumor invades the serosa of the corpus uteri and/or adnexa
IIIb Vaginal and/or parametrial involvement
IIIc Metastases to pelvis and/or paraaortic lymph nodes
IIIcl Positive pelvic nodes
IIIc2 Positive paraaortic lymph nodes with or without positive pelvic lymph nodes
Stage IV a Tumor invades bladder and/or bowel mucosa and/or distant metastases
IVa Tumor invasion of bladder and/or bowel mucosa
IVb Distant metastases, including intraabdominal metastases and/or inguinal lymph nodes

a Includes grades I, II, or III.

b Endocervical glandular involvement only should be considered as stage I and no longer as stage II.

c Positive cytology has to be reported separately without changing the stage.

Stage of disease: Depth of invasion, cervical involvement, adnexal involvement, and nodal metastasis

Staging of patients with malignancies is designed to have prognostic value by classifying the extent of tumor. The survival rate in regard to stage of disease has been consistent, and Table 5.6 and Fig. 5.11 show the 5-year survival rate reported by FIGO (FIGO 1988 staging). The pattern of disease spread in endometrial cancer was evaluated in a prospective study performed by the GOG (GOG protocol 33) and reported by Creasman and colleagues. This study is required reading for physicians who care for patients with endometrial cancer. GOG protocol 33 was a surgical-pathologic study of 621 patients with clinical stage I endometrial cancer who were uniformly treated with total abdominal hysterectomy (TAH), bilateral salpingo-oophorectomy (BSO), peritoneal cytologic evaluation, and selective pelvic and paraaortic lymphadenectomy. Before this study, it was presumed most patients with endometrial cancer were at risk for nodal metastases, and nearly all patients required some form of pelvic radiation therapy (either preoperatively or postoperatively). The study demonstrated important relationships between pathologic factors and risk of nodal disease. For example, factors associated with nodal disease included higher tumor grade, deeper myometrial invasion, cervical involvement, (+) cytology, and LVSI. Because of this study, FIGO accepted a surgical-based staging system, including lymphadenectomy, of endometrial cancer in 1988.

TABLE 5.6
5-Year Survival in Endometrial Cancer: Surgical Stage
From Pecorelli S, editor: FIGO annual report, years 1996–1998, Int J Gynecol Obstet 83:95, 2003.
Stage Patients ( n ) 5-Year Survival (%)
Ia 1063 91
Ib 2735 90
Ic 1219 81
IIa 364 79
IIb 426 71
IIIa 484 60
IIIb 73 30
IIIc 293 52
IVa 47 15
IVb 160 17

Figure 5.11, Carcinoma of the corpus uteri for patients treated in 1990 to 1992 (International Federation of Gynecology and Obstetrics). Survival by surgical stage ( n = 5694).

For endometrial cancer, FIGO staging reflects the progression of disease within the uterus and at extrauterine sites. Approximately 75% of patients with endometrial cancer present with disease limited to the uterus ( Table 5.7 ). For stage I disease, depth of tumor invasion into the myometrium is an important prognostic factor. The degree of myometrial invasion is a consistent indicator of tumor virulence ( Fig. 5.12 ) . DiSaia and associates noted that recurrences were directly related to depth of myometrial invasion in patients with stage I cancer treated primarily with surgery ( Table 5.8 ). The Annual Report of FIGO demonstrated a decrease in the survival rate as myometrial penetration increased ( Table 5.9 ). Lutz and coworkers determined that the depth of myometrial penetration was not as important as the proximity of the invading tumor to the uterine serosa. Patients whose tumors invaded to within 5 mm of the serosa had a 65% 5-year survival rate, but patients whose tumors were more than 10 mm from the serosa had a 97% survival rate. The depth of myometrial invasion is associated with the other prognostic factors, such as tumor grade. As noted by DiSaia and associates, the survival rate of patients with poorly differentiated lesions and deep myometrial invasion is poor in contrast to that of patients who have well-differentiated lesions but no myometrial invasion. This suggests that virulence of the tumor may vary considerably, and as a result, therapy should depend on the combination of prognostic factors.

TABLE 5.7
Distribution of Endometrial Carcinoma by Stage (Surgical)
From Pecorelli S, editor: J Epidemiol Biostat 6:47, 2001.
Stage Patients (%)
I 3839 (73)
II 574 (11)
III 694 (13)
IV 166 (3)

Figure 5.12, Risk assignment based on surgical staging and extent of disease in patients with endometrial cancer.

TABLE 5.8
Relationship Between Depth of Myometrial Invasion and Recurrence in Patients With Stage I Endometrial Carcinoma
Modified from DiSaia PJ, Creasman WT, Boronow RC, et al: Risk factors in recurrent patterns in stage I endometrial carcinoma, Am J Obstet Gynecol 151:1009, 1985.
Endometrial only 7/92 (8%)
Superficial myometrium 10/80 (13%)
Medium myometrium 2/17 (12%)
Deep myometrium 15/33 (46%)

TABLE 5.9
Relationship Between Depth of Myometrial Invasion and 5-Year Survival Rate (Stage I)
From Pecorelli S, editor: FIGO annual report, years 1996–98, Int J Gynecol Obstet 83:95, 2003.
Stage Patients 5-Year Survival (%)
IaGI 698 93
IbGI 1030 88
IcGI 442 87
IaGII 229 91
IbGII 1307 93
IcGII 485 84
IaGIII 66 75
IbGIII 280 82
IcGIII 247 66

Location of the tumor within the endometrial cavity is important because tumors low in the cavity may involve the cervix earlier than fundal lesions. The prognosis for women with cervical stromal involvement (stage II) is worse than those with stage I disease. Cervical involvement is often a surrogate marker for extrauterine disease spread or for risk of local recurrence. Data from GOG 33 showed that those with disease of the lower uterine segment had a higher incidence of pelvic lymph node metastases (16%) than did those with only fundal disease (8%). There is a similar frequency of paraaortic nodal metastases: a 16% incidence from disease of the lower uterine segment and a 4% incidence when only fundal disease is present. Previously, endocervical curettage (ECC) was commonly used to determine whether the patient had cervical involvement. Many false-positive results occur with this technique, however. In addition, the prognostic significance of endocervical glandular spread (formerly stage IIa) has been challenged. The new surgical staging adopted by FIGO in 2009 includes only patients with cervical stromal invasion in the stage II category. In some cases, cervical biopsy or ECC is required in the pretreatment planning of a patient with suspected cervical involvement.

Patients with stage III or IV disease may have a heterogeneous mix of disease characteristics. It is well recognized that endometrial cancer can and frequently does metastasize to the adnexa. Patients with adnexal or serosal involvement are categorized as having stage IIIA disease and are considered to have a higher risk of peritoneal recurrence. In GOG 33, 5% of patients had adnexal involvement. Adnexal metastasis was significantly associated with involved pelvic (32%) and paraaortic (20%) lymph nodes. In a large surgical trial comparing laparoscopy with open staging (GOG Lap 2 trial), 5% of the 2616 patients enrolled were found to have stage IIIa disease (FIGO 1988 staging). In an analysis of 222 patients with clinical stage I carcinoma of the endometrium reported by DiSaia and colleagues, 16 (7%) were found to have metastasis in the adnexa. This finding correlated with many but not all of the other prognostic factors. Spread to the adnexa did not seem to be related to the size of the uterus. The grade of the disease did not appear prognostically important in regard to this in that 6% of patients with grade I tumors had adnexal disease compared with only 10% if poorly differentiated carcinoma was present. The depth of invasion did appear to be significant, however, in that 4% of patients with only the endometrium involved had adnexal spread compared with 24% who had adnexal metastases if deep myometrial invasion was present. If tumor was limited to the fundus of the uterus, only 5% of patients had disease in the adnexa; however, if the lower uterine segment or the endocervix was involved, one-third had spread to the adnexa. When metastasis was present in the adnexa, 60% of patients had malignant cells in the peritoneal cytologic fluid compared with only 11% if the adnexa were not involved. Recurrences appeared in only 14% of these individuals who did not have metastasis to the adnexa compared with recurrences in 38% of patients with adnexal metastasis.

Metastatic spread to lymph nodes (stage IIIc) has prognostic and therapeutic implications. It is clear that lymph node dissection, to date, is the most sensitive way to identify nodal disease. The GOG 33 study demonstrated that as the depth of tumor invasion or tumor grade increased, the frequency of pelvic and paraaortic nodal metastases also increased ( Tables 5.10 and 5.11 ). For all 621 patients, 58 (9%) had positive pelvic nodes, and 34 (6%) had positive paraaortic nodes. Patients with grade I, II and III tumors had pelvic nodal metastases of 3%, 9%, and 18% respectively. Similarly, patients with no myometrial invasion, inner one-third, middle one-third, and outer one-third invasion had 1%, 5%, 6%, and 25% pelvic nodal disease, respectively. The highest risk group included patients with grade III tumors and outer one-third invasion who had pelvic nodal involvement in 34% of cases ( Table 5.12 ). Without a lymph node dissection, one may grossly estimate the probability of nodal involvement based on these data to select for or against the use of adjuvant therapy. This strategy potentially results in undertreatment or overtreatment of patients, however. In two large prospective randomized trials, routine lymphadenectomy resulted in the identification of more patients with nodal disease (9% to 13%) compared with when only clinically suspicious nodes were removed (1% to 3%). It is clear that patients with nodal disease have a poorer prognosis (3- to 5-year survival rate of 50% to 75%) and different patterns of failure (nodal, distant) than patients with negative nodes (3- to 5-year survival rate of 80% to 95%, vaginal cuff failures predominate). Patients with nodal involvement include those with pelvic nodal disease (IIIc1) or paraaortic involvement (IIIc2). The sub-staging was created in 2009 given the belief of different outcomes associated with different levels of nodal involvement. The number of involved nodes and the extent of resection of grossly involved nodes also affect outcome. In addition, Mariani and McMeekin have suggested that patients with positive nodal disease plus other stage III defining features (adnexal or serosal involvement) have a much poorer prognosis than those with nodal disease only. Patients with stage IV or distant disease spread (intraperitoneal, lung, liver) have a particularly poor prognosis, with 5-year survival rate of less than 20%. The extent of surgical resection has been suggested to alter the prognosis in patients with advanced-stage disease. As with ovarian cancer, the extent of resection of disease reflects the biology of the tumor, aggressiveness of the surgery, and response to postoperative therapies.

TABLE 5.10
Grade Versus Positive Pelvic and Aortic Nodes
Modified from Creasman WT, Morrow CP, Bundy BN, et al: Surgical pathologic spread patterns of endometrial cancer: a Gynecologic Oncology Group study, Cancer 60:2035, 1987.
Grade ( n ) Pelvic Nodes (%) Aortic Nodes (%)
GI (180) 5 (3) 3 (2)
GII (288) 25 (9) 14 (5)
GIII (153) 28 (18) 17 (11)

TABLE 5.11
Maximal Invasion and Node Metastasis
Modified from Creasman WT, Morrow CP, Bundy BN, et al: Surgical pathologic spread patterns of endometrial cancer: a Gynecologic Oncology Group study, Cancer 60:2035, 1987.
Maximal Invasion ( n ) Pelvic Nodes (%) Aortic Nodes (%)
Endometrium only (87) 1 (1) 1 (1)
Superficial muscle (279) 15 (5) 8 (3)
Intermediate muscle (116) 7 (6) 1 (1)
Deep muscle (139) 35 (25) 24 (17)

TABLE 5.12
Frequency of Pelvic and Paraaortic Nodal Disease a
Modified from Creasman WT, Morrow CP, Bundy BN, et al: Surgical pathologic spread patterns of endometrial cancer: a Gynecologic Oncology Group study, Cancer 60:2035, 1987.
Depth of Invasion GRADE
Grade I ( n = 180) Grade II ( n = 288) Grade III ( n = 153)
Endometrium only ( n = 86) 0%/0% 3%/3% 0%/0%
Inner third ( n = 281) 3%/1% 5%/4% 9%/4%
Middle third ( n = 115) 0%/5% 9%/0% 4%/0%
Outer third ( n = 139) 11%/6% 19%/14% 34%/23%

a Pelvic nodal positivity/paraaortic nodal positivity.

Tumor grade

The degree of histologic differentiation (tumor grade) of endometrial cancer is a sensitive indicator of prognosis and is included in FIGO stage assignment. The Annual Report on the Results of Treatment in Gynecological Cancer has evaluated survival in regard to grade in patients with clinical stage I AC of the endometrium ( Table 5.13 ). Tumor grade is inversely related to survival; as grade increases, survival decreases. In their review of 244 patients with stage I disease, Genest and colleagues noted that patients with grade I disease had a 5-year survival rate of 96%. This dropped to 79% and 70% for grade II and grade III, respectively. Data presented by Morrow for outcomes in patients enrolled on GOG 33 show that recurrence-free survival markedly diminishes with increasing grade, with progression-free survival (PFS) at 48 months being approximately 95%, 85%, and 68% for grade I, II, and III tumors, respectively. Grade of tumor also correlates with other factors of prognosis. Table 5.14 shows the relationship between differentiation of the tumor and depth of myometrial invasion as reported by Creasman from the GOG 33 study. As the tumor becomes more poorly differentiated, the chances of deep myometrial involvement increase. However, exceptions can occur: Patients with a well-differentiated lesion can have deep myometrial invasion, but patients with a poorly differentiated malignant neoplasm might have only endometrial or superficial myometrial involvement.

TABLE 5.13
Relationship Between Tumor Differentiation and 5-Year Survival Rate, Stage I (Surgical)
From Pecorelli S, editor: FIGO annual report, years 1996–98, Int J Gynecol Obstet 83:95, 2003.
Grade Survival ( n = 5017) (%)
I 91
II 90
III 81

TABLE 5.14
Correlation of Differentiation and Myometrial Invasion in Stage I Cancer
Modified from Creasman WT, Morrow CP, Bundy BN, et al: Surgical pathologic spread patterns of endometrial cancer: a Gynecologic Oncology Group study, Cancer 60:2035, 1987.
Myometrial Invasion GRADE
I (%) II (%) III (%)
None 24 11 11
Superficial 53 45 35
Mid 12 24 16
Deep 10 20 42

Lymphovascular space involvement

Hanson and colleagues described 111 patients with stage I endometrial cancer and found capillary-like space (CLS) involvement in 16. This was most frequently found in patients with poorly differentiated tumors with deep invasion. These patients had a 44% recurrence rate compared with 2% if the CLS was not involved. This was an independently significant prognostic factor. In the GOG study of 621 patients, it was shown that 93 (15%) had CLS involvement. The incidences of pelvic and paraaortic node metastases were 27% and 19%, respectively. This compares with a 7% occurrence of pelvic node metastasis and a 3% occurrence of paraaortic node metastasis when there is no CLS involvement. In risk models predicting risk of recurrence in node-negative, early-stage endometrial cancer, the GOG has suggested that LVSI is an important risk factor.

In the Post-Operative Radiation Therapy in Endometrial Cancer (PORTEC) trial, LVSI was considered a factor associated with distant site of failure. In a pooled analysis of the PORTEC-1 and 2 trials, including 926 tumor samples, substantial LVSI using a 3-tiered approach, had the strongest impact on the risk of distant metastases (HR 4.5; 95% CI 2.4 to 8.5). Furthermore, in multivariate analysis (including prognostic variables such as age, depth of invasion, grade, and treatment), substantial LVIS remained the strongest independent prognostic factor for pelvic recurrence (HR 6.2; 95% CI 2.4016), distant metastases (HR 3.6; 95% CI 1.9 to 6.8), and overall survival (OS; HR 2.0; 95% CI 1.3 to 31) ( ).

Tumor size

Schink and coworkers evaluated tumor size in 91 patients with stage I disease. The incidence of lymph node metastases in patients with tumor size less than 2 cm was only 6%. If the tumor was larger than 2 cm in diameter, there were nodal metastases of 21% and up to 40% if the entire endometrium was involved. Patients with lesions greater than 2 cm in size and less than half myometrial invasion had no nodal metastasis. Using multivariate analysis, the authors showed that tumor size was an independently significant prognostic factor. Watanabe and associates did not find cancer size was predictive of lymph node metastasis. Gynecologic oncologists from the Mayo Clinic have developed a risk model predicting nodal metastases and have suggested that grade I and II tumors that are smaller than 2 cm are particularly low risk for nodal disease.

Peritoneal cytology

The importance of peritoneal cytology in endometrial cancer is controversial. In GOG 33, 76 patients (12%) had malignant cells identified by cytologic examination of peritoneal washings. Of these patients, 25% had positive pelvic nodes compared with 7% of patients in whom no malignant cells were found in peritoneal cytologic specimens ( P > .0001). It is true that peritoneal cytology, to a certain degree, mimics other known prognostic factors—that is, if peritoneal cytologic specimens are positive, other known poor prognostic factors may also be identified. In addition, data from GOG 33 also suggested that the RR for recurrence with positive cytology was 2.4 ( P = .02).

Given the relationship of positive cytology with other known risk factors, it is important to evaluate data sets coming from patients who undergo complete surgical staging. Saga and colleagues reported on a series of 307 patients (32 with positive cytology, 275 with negative cytology) with endometrioid-type cancer, all who underwent complete staging and had negative lymph nodes. The authors reported that 5-year survival rate was 87% with positive cytology compared with 97% with negative cytology. Most patients with positive cytology received chemotherapy postoperatively. Havrilesky et al. reported on experience from Duke University. Patients with positive cytology alone ( n = 37) were compared with patients with adnexal or serosal involvement ( n = 20). The 5-year survival rate was similar between the groups (62% positive cytology, 68% adnexal or serosal disease), but in a multivariable analysis, cytology was an independent predictor of recurrence and poorer survival. This study included patients with non-endometrioid histologies, and lymphadenectomy was not routinely performed. In another review of the literature, Wethington demonstrated that in endometrioid-type tumors with low risk factors (superficial invasion, grade I or II) the recurrence rate was only 4%. By comparison, 32% of patients with higher-grade tumors or deeper invasion and positive cytology had a recurrence. These results suggest that for at least a subset of cytology patients, the risk of recurrence approximates that driven by other uterine factors (depth of invasion, tumor grade). In 2009, FIGO dropped cytology as a stage IIIa defining characteristic.

Milosevic and colleagues reviewed 17 studies. In 3820 patients, the prevalence of positive cytologic findings was 11%. The three largest studies totaling more than 1700 patients (Haroung and associates, Turner and colleagues, Morrow and coworkers) using multivariate analysis noted that the finding of malignant cells on cytologic examination was independently significantly associated with either recurrence or reduced survival. Pooled odds ratio for the entire series was 4.7 (CI 3.5 to 6.3) for disease recurrence. All studies note the highest correlation of malignant cytologic specimens with extrauterine disease. An alternate observational retrospective cohort analysis of 16,851 women using the National Cancer Database (NCDB) similarly found positive cytology to be associated with compromised 4-year OS in patients with stage I to II disease ( ). Additional studies have confirmed these findings ( ). It does appear that with multivariate analysis, the presence of malignant cells is an important prognostic factor even when disease is limited to the uterus in all histologic subtypes, although as noted previously, it is no longer incorporated into the FIGO staging algorithm.

Optimal therapy has not been determined to date. Historically, intraperitoneal 32 P or oral progestins have been used to manage disease in patients with positive cytology. Today, uterine risk factors are largely used to define postoperative adjuvant therapy independent of cytology status and, increasingly, in otherwise low-risk patients (endometrioid-type tumors, negative nodes), observation is often considered. Chemotherapy has also been used in the adjuvant setting for patients with positive cytology, with retrospective studies suggesting a potential benefit.

Molecular indices

Hormone receptors.

Historically, estrogen and progesterone receptors were the first “targets” in the molecular biology of endometrial cancer. Using multivariate analysis to analyze hormone receptor status, Creasman and associates noted that in stage I and stage II cancers, progesterone receptor–positive status was a highly significant, independently prognostic factor in endometrial cancer. Without progesterone receptor status in the model and with the evaluation of estrogen receptor status, estrogen receptor–positive status was an independent prognostic factor but not to the degree of progesterone receptor–positive status. Hormone receptor status may closely mirror grade, however.

Thankfully, we have seen a transition from disease homogeneity to molecular granularity in the endometrial cancer space, with an explosion of research into the molecular makeup of this disease. Cytogenetic studies have described gross chromosomal alterations, including changes in the number of copies of specific chromosome. The extent of abnormalities in a given tumor is relatively low. About 80% have normal diploid DNA content. Aneuploidy in 20% is usually associated with high-grade, extrauterine disease, high-risk histology, and poor prognosis. So-called loss of heterozygosity occurs at a relatively low frequency compared with other solid tumors. When chromosomal loss of heterozygosity does occur, underlying molecular genetic defects have been observed on 17p and 10q that correlate with mutational inactivation of TP53 and PTEN, respectively. Individual tumors with a greater number of gains and losses are associated with a poorer prognosis, and some changes seen in cancer are also present in atypical hyperplasia but not simple hyperplasia.

Mutational activation or aberrant expression of some oncogenes has been described but to a lesser degree than tumor suppressor genes. The RAS gene family is the most commonly identified oncogene aberration in human cancers and is present in 10% to 30% of endometrial cancers. This mutation appears to occur early in the neoplastic process, and the incidence is the same in endometrial hyperplasia. Correlation of RAS mutation to survival has produced conflicting results. About 10% to 15% of endometrial cancer overexpress ERBB2 (HER2/neu). Overexpression appears to be confined to high-grade or advanced-stage tumors. More specifically ERBB2 amplification is most common in USC, occurring in 25% to 45% of these tumors, although estimates of overexpression range from 14% to 80% ( , , ). This significant variation is likely related to methodology, criteria used to define overexpression, and interpretation. More recently, a multi-institutional cohort study of 169 patients with stage 1 USC identified that HER2 overexpression (26% HER2-positive; defined as 3+ IHC of fluorescent in situ hybridization [FISH] positive), was associated with a significantly higher risk of disease recurrence, and compromised PFS and OS in multivariate models, despite similar rates of adjuvant therapy with carboplatin and paclitaxel (Erickson, 2020, #2598).

The FMS oncogene encodes a tyrosine kinase, which serves as a receptor for macrophage colony-stimulating factor (m-CSF). Expression of FMS correlates with advanced-stage, high-grade, and deep myometrial invasion. Expression of C-MTC, which has been observed in normal endometrium and endometriosis, has a higher expression in secretory endometrium. Several studies suggest amplification is present in a fraction of endometrial cancers.

Mutation of TP53 tumor suppressor gene, the most common genetic abnormality currently recognized in human cancers, is present in 10% to 30% of endometrial cancers. Overexpression or mutation (or both) is associated with prognostic factors. In a study of more than 100 endometrial hyperplasia specimens, TP53 mutation was not present. PTEN mutation analysis in endometrial cancer indicates that this gene is somatically inactivated in 30% to 50% of all tumors, the most frequent molecular genetic alteration defined in endometrial cancer. There does appear to be a correlation between microsatellite instability (MSI) and PTEN mutation. PTEN mutation is observed in 20% of endometrial hyperplasia, suggesting that this is an early event in the development of some type I endometrial cancers.

Inherited mutations in gene encoding DNA mismatched repair proteins, including MLH1, PMS2, MSH2, and MSH6, are responsible for HNPCC, for which endometrial cancer is the second most common cancer in women with these mutations. Cancers in these individuals are characterized by frame shift mutations in multiple microsatellite repeat sequences throughout the genome. This mismatch repair deficiency (dMMR) is seen in approximately 20% to 30% of sporadic endometrial cancers, the vast majority of which are endometrioid histology. In these sporadic cancers, acquired mutation in mismatched repair genes is rare, although epigenetic silencing of MLH1 can be seen. Endometrial cancers that exhibit dMMR tend to be type I. dMMR can be identified in some cases of complex hyperplasia associated with endometrial cancer but is not seen in USC, and uncommonly in clear cell uterine cancer. Importantly, dMMR, which is defined using immunohistochemistry, does not equate to MSI, which is determined using PCR-based assessments of five microsatellite markers. Cancer specimens with two or more of the five markers exhibiting instability are defined as MSI-high based on the National Cancer Institute Workshop guidelines ( ).

Type I endometrial cancers are commonly described to include tumors seen in obese and nulliparous women, are well differentiated, are superficially invasive, and frequently carry a good prognosis. These tumors also share several common molecular changes and tend to have the following genetic features: diploid, low allelic imbalance, MLH1 methylation, KRAS, and PTEN mutations. In contrast, type II endometrial cancers are associated with poor prognostic pathologic features, have aneuploid, high allelic imbalance, KRAS, TP53, and HER2/neu changes.

The Cancer Genome Atlas (TCGA) Research Network has further improved our understanding of the molecular characteristics of endometrial cancer following the publication of a pivotal paper describing molecular alterations identified in endometrial cancer samples ( ). A total of 373 endometrial cancer samples were evaluated using array- and sequencing-based technologies, including low-grade and high-grade endometrioid histologies as well as serous uterine carcinomas. The authors described four molecular groups, including (1) POLE (ultra-mutated) tumors (~7%), (2) microsatellite unstable tumors (hypermutated, MSI-high mostly as a result of MLH1 promoter methylation) (~28%), (3) copy-number low tumors, mostly endometrioid histology (~39%), and (4) a copy-number high (serous-like) cohort with frequent P53 mutations (26%). POLE -mutant tumors were found to have significantly better PFS, while copy-number high tumors were associated with the worst prognosis. The TCGA additionally confirmed the frequency of PI(3)K/AKT pathway mutations, which occurred more frequently in endometrioid endometrial cancers (93%), than any other tumor type examined by the TCGA. In the TCGA analysis, almost all USC cases clustered into a mitotic transcriptional profile, with evidence of mRNA changes associated with cell cycle dysregulation, including in CCNE1 , PIK3CA, MYC , and CDKN2A . Although informative and clinically relevant, the data generated in the TCGA necessitate use of data-intensive and cost-prohibitive methodology.

Alternate pragmatic and potentially generalizable methods have been developed to allow for patient classification similar to the initial TCGA publication. Talhouk and colleagues were successful in replicating the molecular classification of the TCGA using the Proactive Molecular Risk Classifier for Endometrial Cancer (ProMisE) which includes: (1) mismatch repair protein IHC, (2) POLE mutational analysis, (3) and p53 IHC as a surrogate for copy-number status ( ). Initial assessment was based on MMR status, with dMMR tumors classified as dMMR and MMR proficient (pMMR) tumors tested for POLE mutations. POLE mutants were classified as POLE exonuclease domain mutations, with POLE wild-type tumors examined for p53 status. pMMR, POLE wild-type tumors were classified as p53 wild-type or p53 abnormal (null/missense mutations). The ProMisE classifier was subsequently validated in a large cohort series, confirming the prior TCGA findings that POLE mutations had the best prognosis and p53 mutants the worst.

The NRG Oncology/GOG 210 study was an observational study that collected tissue, serum, and urine on over 5500 endometrial cancer patients, correlated with clinical and pathologic data. This dataset has resulted in numerous publications and continues to identify relevant molecular, clinical, and prognostic factors in endometrial cancer care. A molecular classification for risk prediction was developed using 982 tumor samples from GOG 210. Investigators assessed MMR defects (MSI, MMR IHC, and MLH1 methylation), POLE mutations, and loss of heterozygosity at microsatellite repeats. Using four classifications (dMMR copy number altered [can], copy number stable, and POLE mutant), the authors were able to stratify patients by PFS and OS. The classification system remained statistically significant in multivariable analyses. CNA had the worst PFS and cancer-specific survival, whereas the POLE group had the best outcomes.

Research continues in an effort to identify molecular markers that may augment clinical-pathologic information in establishing risk and informing adjuvant therapy. Other cancer types (breast, prostate, bladder) have well-validated disease-specific nomograms predicting risk and benefit to adjuvant therapy. Similarly developed and validated models for endometrial cancer are needed. Emerging data in the endometrial cancer space include the PORTEC-3 molecular classification for high-risk endometrial cancer ( ). Investigators looked to examine prognosis and impact of chemotherapy for predefined molecular subgroups (abstracted from the TCGA endometrial) using 423 patient samples, 103 of which had stage IIIc disease. Submitted tissue specimens were evaluated using p53 IHC, MMR IHC, as well as POLE exonuclease domain DNA sequencing. Samples were then classified as p53 abnormal, POLE -ultramutated, dMMR, or no specific molecular profile (NSMP). Five-year recurrence free survival (RFS) was 48% for patients with p53abn EC, 98% for POLE- mutated EC, 72% for dMMR EC, and 74% for NSMP EC ( P < .001). The 5-year RFS with CTRT versus RT for p53abn EC was 59% versus 36% ( P = .019); 100% versus 97% for patients with POLE -mutated EC ( P = .637); 68% versus 76% ( P = .428) for dMMR EC; and 80% versus 68% ( P = .243) for NSMP EC. The application of the predefined molecular subgroups to the PORTEC-3 dataset suggest a potential predictive capacity of EC molecular classification in identifying the benefit of chemotherapy over radiation. The ongoing PORTEC-4a trial looks to capitalize on these findings by comparing standard adjuvant brachytherapy with individualized treatment algorithms based on integrated molecular profiling. Furthermore, the suggestion of limited benefit of adjuvant chemotherapy in the dMMR cohort enrolled on PORTEC-3 catalyzed the development of novel trials examining adjuvant immune checkpoint inhibitors following radiation versus radiation alone.

Relevant Biomarkers in Endometrial Cancer and Treatment Options
Endometrial Cancer Biomarker Potential Treatment Implications
HER2-positive (defined as 3+ IHC of FISH positive) Incorporation of anti-HER2 treatments: Trastuzumab
Mismatch repair deficiency (dMMR) Immune checkpoint inhibition: Pembrolizumab
POLE mutated Immune checkpoint inhibition: Pembrolizumab
TMB-high (>10 mutations/Mb) Immune checkpoint inhibition: Pembrolizumab
Estrogen and Progesterone receptor positive Hormonal Therapy (monotherapy or combination regimens)
FISH, Fluorescent in situ hybridization; IHC, immunohistochemical.

Correlation of multiple prognostic factors

GOG protocol 33 detailed the extent of disease identified at surgery and correlated this with outcomes. Although single factors were associated with recurrence, the combination of factors could also establish risk. For example, Morrow showed that patients with no risk factors (LVSI, cervical involvement, adnexal involvement, nodal disease) had a very low risk of recurrence, but 20% with one, 43% with two, and 63% with three or more factors recurred. In multivariate analysis, patients with disease limited to the uterus were at increased risk for recurrence if there were deep myometrial invasion, vascular space involvement, or positive washings.

Data from three randomized trials evaluating the use of postoperative pelvic radiation therapy have been useful in shaping models predicting risk of recurrence after surgery. Aalders identified a subset of the 95 of 540 (18%) patients enrolled in a randomized trial of vaginal cuff brachytherapy (VCB) with or without pelvic radiation therapy with grade III tumors and any myometrial invasion to be particularly notable for risk of recurrence. Patients with these factors had a pelvic recurrence rate of 20% with vaginal brachytherapy alone compared with 5% with the combination of pelvic and vaginal radiation. It is interesting that approximately 15% of patients with these tumor characteristics failed at a distant site, regardless of radiation technique. The PORTEC trial compared pelvic radiation therapy with observation after hysterectomy without lymph node dissection in patients with endometrial cancer. In this trial of 714 patients, a subset of patients with grade III tumors (10%), age older than 60 years (72%), or greater than 50% depth of invasion (59%) was identified in which having two of three factors defined a high-risk group. The locoregional failure rate was 23% in this group who underwent surgery followed by observation compared with 5% after pelvic radiation therapy. There was no difference in distant sites of recurrence (~5% each group) or in cancer-related deaths (8% observation, 11% radiation). The GOG conducted a randomized trial (GOG 99) of observation versus pelvic radiation in a group of patients with surgically documented negative nodes and any amount of myometrial invasion. Depending on patient age and number of risk factors (LVSI, grade I or II tumor, outer one-third myometrial invasion) ( Table 5.15 ), a high-intermediate (H-IR) risk group could be identified. The H-IR group accounted for one-third of all patients enrolled but two-thirds of recurrences. Perhaps equally important was the identification that two-thirds of the patients enrolled who did not have H-IR features had an incredibly low risk of recurrence (2% to 3%), suggesting that in addition to defining high-risk groups, low-risk groups who may avoid postoperative adjuvant therapy may also be identified.

TABLE 5.15
High-Intermediate Risk Model: Early-Stage Endometrial Cancer
Modified from Keys HM, Roberts JA, Brunetto VL, et al: A phase III trial of surgery with or without adjunctive external pelvic radiation therapy in intermediate risk endometrial adenocarcinoma: a Gynecologic Oncology Group study, Gynecol Oncol 2004;92:744–751.
Age Risk Factor
Any age + 3 factors LVSI
≥50 years + 2 factors Grade II or III tumor
≥70 years + 1 factor Outer one-third invasion
LVSI , Lymphovascular space invasion.

The value of risk models is that they can be applied to an individual patient incorporating all of the information (age, uterine characteristics) to provide a reasonable estimate of risk of recurrence and probability of benefit of selected adjuvant therapy.

Treatment

Surgical management of endometrial cancer

Sampling of the endometrium in symptomatic postmenopausal patients is recommended as the first diagnostic technique. If histologic findings are “negative,” the patient is observed. An endometrial curettage is recommended if the patient continues to be symptomatic after the negative endometrial biopsy result. After a tissue diagnosis of endometrial cancer is established, the patient should be assessed for surgical options of therapy. Most patients require routine complete blood counts (CBC), metabolic profile, and a metastatic evaluation with chest radiography. Routine use of computed tomography (CT) or MRI scans has not shown to be useful and should be reserved for patients with high-risk tumor types, evidence of intraperitoneal disease, positive chest radiograph, or clinical evidence of metastatic disease. Most patients with endometrial cancer are candidates for definitive surgery, including surgical staging. Routes of hysterectomy include vaginal, abdominal, laparoscopic (total or assisted), and robotic and have broadened the surgical options for patients.

The surgical evaluation of most patients with endometrial cancer requires a thorough inspection of the peritoneal cavity, collection of cytologic washings, and sentinel lymph node (SLN) dissection in addition to hysterectomy. Cytologic evaluation of peritoneal fluids, or washings, has been a common surgical step in staging endometrial cancer because of associations with extrauterine disease and prognosis. After the peritoneal cavity is opened, an assessment of the amount of peritoneal fluid in the pelvis is made. If none is present, approximately 100 mL of normal saline solution is injected into the pelvis and aspirated. This can be done with the suction irrigation device at the time of minimally invasive surgical staging. Peritoneal cytologic evaluation may be performed in patients undergoing surgery for endometrial cancer, although FIGO no longer uses cytology as a stage-defining characteristic. Omental biopsy may be considered in patients with gross spread to the omentum or adnexa or in cases with high-risk histologies such as serous or clear cell tumors. In a large GOG study of 621 staged patients, 6% of patients with clinical stage I disease were noted to have intraperitoneal disease.

The definitive treatment for patients with endometrial cancer is hysterectomy. The hysterectomy should be extrafascial, and removal of the upper vagina does not appear to decrease vault recurrences ( Fig. 5.13 ). Extirpation of the uterus removes the primary tumor and can provide important information that can be used to estimate risk of spread to lymph nodes or risk of recurrence. Removal of the adnexa is thought to be important given that approximately 5% of endometrial cancers have metastatic disease to the ovaries, fallopian tubes, or both. In addition, synchronous ovarian and endometrial cancers are not infrequent, particularly in younger patients. Historically, total hysterectomy and BSO have been the hallmarks of therapy for endometrial cancer. There are no data to suggest that the route of hysterectomy influences RFS or OS outcomes for patients; as such, the approach (robotic, laparoscopic, or abdominal) that is the safest and facilitates rapid recovery is recommended. Minimally invasive surgical approaches are preferred given the reduced morbidity and rapid recovery without decrement in oncologic outcome, as supported by the LAP-2 publications.

Figure 5.13, Total abdominal hysterectomy and bilateral salpingo-oophorectomy showing large polypoid adenocarcinoma of the endometrium with deep myometrial invasion.

Several studies note the role of vaginal hysterectomy in highly selected patients with endometrial cancer for whom surgical staging cannot be performed safely using alternate approaches (significant cardiopulmonary medical comorbidities, advanced age). Because factors associated with the choice of vaginal hysterectomy (morbid obesity, medical comorbidities) are often associated with favorable uterine characteristics (low-grade lesions, smaller uterus), it is not surprising that survival rates are comparable to those of the abdominal approach. Chan and coworkers reviewed 51 medically compromised patients treated with vaginal hysterectomy and reported 3- and 5-year disease-specific survival rates of 91% and 88%, respectively. Of note, approximately 50% of women could not have the ovaries removed at the time of vaginal hysterectomy . Assessment of lymph nodes is also not feasible with the vaginal approach and should be a component of patient counseling. Smith evaluated 63 patients with obesity or medical comorbidities and found that vaginal hysterectomy was safe and well tolerated in this patient population. Vaginal hysterectomy may represent a reasonable option for patients who may not tolerate other approaches or for whom surgical staging is not being considered (atypical hyperplasia, some patients with grade I cancers).

Over time, there has been a more widespread use of lymphadenectomy in the management of patients with endometrial cancer. Lymph node dissection provides the best estimate of spread of disease (vs. palpation or use of imaging studies), the lymph node status is prognostically important (as evident from the different survival rates seen with different stages of disease), and patients who are found to have positive or negative nodes receive different postoperative therapy versus patients with unknown status of lymph nodes. This may be particularly relevant in the context of recent clinical trials suggesting survival benefits with incorporation of adjuvant chemotherapy in patients with node positive endometrial cancer.

Unfortunately, complete pelvic and/or paraaortic lymphadenectomy is associated with intraoperative and postoperative complications including nerve and vascular injury, lymphocyst formation, lymphedema, and neuralgia and is difficult to accomplish in morbidly obese patients ( ; ). Furthermore, two large prospective phase III trials failed to show a survival advantage with incorporation of lymph node dissection into the surgical staging algorithm.

In a large multicentric study (A Study in the Treatment of Endometrial Cancer [ASTEC]), 1408 patients were randomly assigned to TAH or BSO with or without pelvic lymphadenectomy (PLA). With a median follow up of 37 months, there was no difference in survival between the two groups. This was an intent-to-treat study and has been criticized as having a relatively large noncompliance in regard to lymphadenectomy and subsequent therapy. In the lymphadenectomy arm, almost half had no nodes or a small number (≤9) removed. This study was further complicated by the fact that many of these patients were secondarily randomly assigned into a postoperative radiation study, not taking into consideration surgical findings. para-aortic lymphadectomy (PALA) was not the standard management, although some patients had these nodes removed.

The second study was published by Panici and colleagues. The Italian study randomly assigned 514 stage I patients to systematic PLA or no lymphadenectomy. Although 13% of patients in the lymphadenectomy arm had known metastasis versus 3% in the no-lymphadenectomy arm, there was no difference in recurrence or OS. The protocol required a minimum of 20 lymph nodes to be removed; however, the lymphadenectomy was limited to the pelvis, although PALA could be removed at the discretion of the surgeon. Postoperative radiation was left to the discretion of the treating physician. Adjuvant therapy was similar in the two groups. Whether or not this had an effect on survival is unknown because this was not standardized.

In an effort to mitigate adverse events associated with comprehensive lymphadenectomy, while recognizing the prognostic value of defining lymph node status, investigators explored the role of sentinel lymph node dissection in the management of patients with endometrial cancer. The initial feasibility of sentinel lymph node dissection in endometrial cancer was described in retrospective, single institution studies and smaller prospective trials ( ; ). The Fluorescence Imaging for Robotic Endometrial Sentinel lymph node biopsy (FIRES) trial was designed to estimate the sensitivity and negative predictive value of sentinel-lymph-node mapping using robotic assisted fluorescence imaging of the tracer indocyanine green (ICG) in detecting lymphatic metastases in patients with endometrial cancer ( Table 5.16 ) ( ). This multicenter, cohort study enrolled a total of 385 patients with endometrial cancer of any histology, and clinical stage 1 disease, without physical examination or radiographic imaging suggestive of extrauterine disease. Investigators injected ICG into the cervix at a dose of 0.5 mg/mL by diluting 1 mL of the stock solution (2.5 mg/mL) into 4 mL of sterile water. A spinal needle was used to inject 1 mL (0.5 mg) of the ICG solution into the uterine cervix at 3 and 9 o’clock of the ectocervix to a 1 cm depth, achieving a total dose of 1 mg. Successful mapping was defined by observing a channel leading from the cervix directly to at least one candidate lymph node in at least one hemi-pelvis. Identified sentinel lymph nodes were then retrieved and labelled for location ( Fig. 5.14 ). Completion bilateral lymphadenectomy (removal of all remaining non-sentinel lymph node tissue within the relevant nodal basins) was then done on all patients according to the GOG surgical handbook. PLA was required in all patients, with PALA at the discretion of the operating physician. Excellent surgical quality control was incorporated into this trial. Pathologists assessed SLNs for metastases by hematoxylin and eosin (H&E) staining, followed by pancytokeratin AE1 and AEs IHC ultra-staging if H&E assessment was negative. Importantly, 102 patients (29%) had high-grade histology (grade III endometrioid, serous and clear cell cancer). Mapping identified at least one SLN in 293 (86%) of 340 patients. Two of three patients with a mapped isolated paraaortic lymph node had metastatic disease, with otherwise negative pelvic LN. They found an impressive 97.2% sensitivity and 99.6% negative predictive value ( ). In this large study, SLN mapping was not successful in 13.8% of eligible patients.

TABLE 5.16
Findings From Three Pivotal Sentinel Lymph Node Clinical Trials
FIRES ( , #2479)
  • n = 385

  • 82% endometroid

  • 12% serous

  • 4% carcinosarcoma

  • 2% clear cell

Successful mapping of at least one SLN in 86% of patients (52% bilateral) Sensitivity 97% NPV 99.6% False negative rate 3%
SENTOR (Cusimano, 2021, #2601)
  • n = 156

  • 42% endometrioid (22.5% grade III)

  • 33% serous

  • 11% carcinosarcoma

  • 2% clear cell

Successful mapping of at least one SLN in 97.4% (77.6% bilaterally) Sensitivity of 96.3% NPV 99.2% False negative rate 3.7%
SHREC (Persson, 2019, #2602)
  • n = 257

  • 23% serous

  • 5% clear cell

  • 5% carcinosarcoma

  • 65% endometrioid (13.3% grade III)

Successful bilateral mapping in 82% (95% with reinjection) Sensitivity of 98% NPV 99.5% False negative rate 3.5%

Figure 5.14, Fluorescence imaging of the tracer indocyanine green (ICG) in detecting sentinel lymph nodes with processing.

Following publication of the FIRES trial, SLN dissection was adopted into the staging algorithm for endometrial cancer (both minimally invasive and open approaches), with the NCCN recognizing SLN dissection as an appropriate surgical option (Category 2A). It should be emphasized that the success of SLN mapping is dependent on appropriate application of the algorithm, which includes completion of side-specific comprehensive lymph node dissection in cases of failed mapping and the removal of any suspicious appearing LN irrespective of mapping. Lastly, in cases where SLN mapping is not successful, providers can use uterine factors to guide and inform need for nodal assessment.

An unanticipated consequence of incorporating SNL dissection into uterine cancer care has been the identification of isolated tumor cells (ITCs) (<0.2 mm) on pathologic nodal evaluation. In the FIRES trial, 19 patients had low-volume disease identified in the SLN, with 10 subjects having ITCs. ITCs do not upstage patients (N0(i+)), and to date, studies suggest no difference in outcome based on choice of adjuvant therapy ( ). In a large, multi-institutional, retrospective study of 175 patients with stage I/II endometrioid endometrial cancer and SLN ITCs, adjuvant therapy did not impact oncologic outcomes (RFS), suggesting observation may be a reasonable approach in this setting (Backes, 2021, #2599). Longer follow-up time and sample sizes are required to confirm these findings.

When comprehensive lymphadenectomy is required, the retroperitoneal spaces in the pelvis are opened in routine fashion. The vessels are outlined, and the lymph node–bearing tissue along the external iliacs from the bifurcation to the inguinal ligament is removed. The obturator fossa anterior to the obturator nerve is cleaned of lymphoid tissue. Lymph nodes along the common iliacs are also removed. The left and right paraaortic nodes are approached by retracting the small intestine into the upper abdomen and incising the peritoneum over the upper common iliac artery and lower aorta. The main vessels are outlined, and the ureter is retracted laterally. On the right, the tissue overlying the vena cava and the aorta are removed en bloc, beginning at the bifurcation of the aorta and extending caudad. On the left, the left common nodes are frequently quite lateral. Using this technique, one should have a total of 20 to 30 pelvic and paraaortic lymph nodes available for histologic evaluation. The upper limit of the dissection (unless enlarged nodes are noted above this area) is usually the inferior mesenteric artery (IMA), but some surgeons suggest that a comprehensive lymphadenectomy should extend to the level of the renal vessels.

In the context of prior studies suggesting two nodal drainage basins—pelvic and paraaortic—the initial adoption of sentinel LN algorithms was accompanied by a concern for paraaortic failures in patients with negative pelvic sentinel LN. Prior studies indicated that when lymph nodes are positive, approximately 50% to 60% of the time paraaortic nodes are involved. In a retrospective study from the Mayo Clinic, 137 patients at high risk for nodal involvement who underwent PALA (PAL+) were compared with those who did not undergo surgical evaluation of the paraaortic nodes (PAL−). The 5-year survival rate was 85% for PAL + patients compared with 77% for PAL patients. In 51 patients with pelvic or paraaortic node metastasis, survival was 77% for PAL + patients compared with 42% in the PAL group. It is generally thought that the rate of isolated paraaortic lymph node metastasis is less than 5%; metastasis isolated to the paraaortic lymph node and metastasis above the IMA are seen. Mariani and colleagues reported on a study of 482 patients with endometrial cancer at the Mayo Clinic. A total of 281 underwent lymphadenectomy to the renal vessels, and 22% had positive lymph nodes; of these, 51% had both positive pelvic and paraaortic nodes, 33% had positive pelvic nodes only, and 16% had isolated involvement of the paraaortic nodes. Of note, 46% of those with isolated paraaortic lymph nodes were only positive above the IMA, and 77% had at least one metastatic node located above the level of the IMA. However, the authors do report that there were no positive lymph nodes detected in those with grade I or II tumors with a tumor size of smaller than 2 cm with less than 50% invasion, raising the question of the benefit of lymphadenectomy in the truly low-risk group. Abu-Rustum and colleagues demonstrated a 1% risk of isolated paraaortic lymph node metastasis in both the low- and high-grade tumors.

In a meta-analysis which included 3536 patients in six studies, investigators showed higher positive pelvic nodal detection rates, no difference in paraaortic nodal detection rates, and no difference in overall recurrence or nodal recurrence rates in SLN compared with complete lymphadenectomy ( ). Specific to high-risk histology, a prospective study examined women with grade III endometrioid, serous, clear cell, and carcinosarcoma by SLN biopsy followed by full pelvic and paraaortic lymphadenectomy; they found 95% sensitivity and 98% negative predictive value and bilateral mapping rates of 58% and unilateral mapping rates of 40%, supporting the use of SLN in high-risk patients ( ).

Since 1988 when FIGO changed endometrial staging from clinical to surgical, the debate as to whether the lymphadenectomy is only diagnostic, which is an important determinate, or whether it could also be therapeutic has persisted. Kilgore and associates, in evaluating 649 patients, noted that those who underwent multiple-site lymph node removal had significantly better survival rates than those patients who had no lymph nodes removed ( Figs. 5.15 and 5.16 ). Lymph node removal resulted in a better survival rate than for those without lymph node removal plus postoperative radiation. Cragun and colleagues reported that patients with grade III or poorly differentiated cancers with more than 11 lymph nodes removed had an improved survival (HR, 0.25) and PFS (HR, 0.26) compared with those with fewer than 11 lymph nodes removed. However, the number of lymph nodes removed was not predictive of survival outcome in those with grade I and II tumors. This association between lymphadenectomy and improved survival remained when controlling for adjuvant radiation treatment, emphasizing the impact of extensive lymph node dissection. A retrospective review by Chan and colleagues of more than 12,000 patients showed an increased 5-year survival rate for the intermediate- and high-risk group that underwent an extensive lymph node dissection. However, there was no benefit of nodal dissection seen in the low-risk group.

Figure 5.15, Survival by nodes sampled and risk groups: multiple-site pelvic node sampling versus no nodes. Low-risk group, P = .026; high-risk group, P = .0006.

Figure 5.16, Survival comparisons of multiple-site pelvic node sampling with whole-pelvic (WP) radiation therapy (RT): multiple nodes without RT versus no nodes plus WP RT. Low-risk group, P = .003; high-risk group, P = .041.

Removal of nodes involved by tumor has been supported as a therapeutic option. In a study reported by Havrilesky et al., 91 patients were identified with stage IIIc disease. There were 39 with microscopic involvement of the lymph nodes and 52 with grossly enlarged nodes. After surgery, 92% received some type of adjuvant therapy with 85% receiving radiation therapy. The survival rate (5 years) was 58% in the 39 with microscopic lymph nodes, 48% in 41 patients with grossly positive lymph nodes completely resected, and only 22% in the 11 with unresected lymph nodes. The authors believed that these data suggested a therapeutic benefit for lymphadenectomy. Similarly, in a small study of 41 patients, Bristow and coworkers noted disease-free survival (DFS) was improved if patients with bulky adenopathy underwent complete resection of involved nodes compared with patients who had gross residual disease in lymph nodes remaining after surgery (37.5 vs. 8.8 months; P = .006).

Onda and colleagues carried out thorough pelvic and paraaortic lymphadenectomies on 173 patients with stage I to III endometrial cancer. The average numbers of lymph nodes removed were 38 pelvic and 29 paraaortic. There were 30 patients (17%) with positive nodes: 10 to pelvis only, two paraaortic only, and 18 with metastasis to both pelvic and paraaortic nodes. Selected patients received radiation therapy with extended fields, combination chemotherapy, or both. In the 10 patients with only pelvic metastasis, the 5-year survival rate was 100%; it was 75% in those with paraaortic involvement. The authors suggest that, although postoperative treatment may contribute to these excellent results, systematic pelvic and PALA was a contributing factor.

One of the important effects of lymph node dissection is that it identifies that most patients with negative lymph nodes are at very low risk for recurrence. As such, lymph node dissection has allowed for modifications of postoperative therapy away from pelvic radiation. In one study, Mohan and associates evaluated 159 stage I patients who had full PLA and received vaginal brachytherapy rather than the more traditional pelvic radiation. In general, vaginal brachytherapy is a quicker therapy (1 to 3 days vs. 5 to 6 weeks for pelvic radiation) and is associated with fewer side effects. In the Mohan study, the 15-year OS was 92%, and the recurrence rate was 4%, all at distant sites. Podratz and associates reviewed four studies that used thorough lymphadenectomies in moderate- and high-risk patients who did not receive postoperative radiation therapy. There were 20 recurrences (7%) in 305 patients; only five recurrences were local or regional, with four being in the vagina. Those four did not receive postoperative brachytherapy but were salvaged with subsequent radiation.

The disadvantages of a lymph node dissection include that the performance of lymph node dissection requires a specially trained surgeon, has the potential to increase complications, as previously discussed, and has not been prospectively validated to improve outcomes. Two phase III trials compared the use of lymph node dissection or not in patients with endometrial cancer with no identifiable improved outcome with lymph node dissection.

The Cochrane Database concluded that there was no overall statistical difference between PFS and OS in patients who had a lymphadenectomy versus those who did not but did report more morbidity for those patients who had a lymphadenectomy. Cochrane only considered the ASTEC and the Panici trial in reaching this conclusion.

Patients with stage II carcinoma of the endometrium, because of extension of disease into the endocervix, will have a greater propensity for lymph node metastasis. For example, in the GOG 33 protocol, 16% of patients with cervical involvement had positive pelvic lymph nodes versus 8% with a fundal location of tumor. Even in patients with occult cervical involvement, 19% of patients had lymph node metastases. In addition, parametrial and vaginal involvement are thought to be more common when the cervix is involved. As such, surgery in the form of radical hysterectomy and lymphadenectomy can be considered when there is gross cervical involvement with tumor. Because many cases of cervical involvement are occult and only recognized after surgery, simple hysterectomy with lymph node sampling (SLN algorithm) appears to be adequate surgery in most cases. Postoperative radiation therapy can be planned, depending on surgical-pathologic findings, including use of pelvic radiation or VCB, both, or neither.

Currently, a minimally invasive approach to endometrial cancer staging is advocated given similar oncologic outcomes with reduced intra- and perioperative morbidity. A large GOG study, led by Walker and colleagues (GOG LAP2), compared laparoscopy versus laparotomy in the surgical management of endometrial cancer. Patients were randomly assigned between laparoscopic versus open laparotomy surgical staging. Laparoscopy was initiated and completed without conversion in 74%. Conversion from laparoscopy to laparotomy was secondary to poor visibility in 15%, metastatic cancer in 4%, and bleeding in 3%. Patients randomly assigned to undergo laparoscopy had significantly fewer moderate to severe postoperative (14% versus 21%) complications and similar rates of intraoperative complications. Length of hospital stay was significantly shorter for those randomized to undergo laparoscopy (median 3 days vs. 4 days), but operative time was significantly longer (median, 204 vs. 130 minutes). Pelvic and paraaortic nodes were removed in 92% of patients undergoing laparoscopy and 96% of patients undergoing laparotomy, and cytology was performed in 96% versus 98%. Neither treatment arm demonstrated an improved ability to detect metastatic disease. Quality-of-life evaluation found a better body image and return to normal activities for the patients undergoing laparoscopy. Laparoscopic surgical staging is feasible and safe for patients with uterine cancer and results in fewer complications and shorter hospital stays. Recurrence rates were lower than anticipated; the estimated 3-year recurrence rate was 11% with laparoscopy and 10% with laparotomy, and the estimated 5-year OS rate was almost identical in both arms (90%).

Today, advancements of laparoscopic surgery, including use of a robotic surgical platform, have permitted minimally invasive approaches with a reduction in conversion rates to open laparotomy. Robotic surgery offers three-dimensional graphics, visual magnification, improved ergonomics, masking of operating physician tremor, and greater dexterity of surgical instruments. Robotic surgery has expanded the application of laparoscopy, especially in obese patients or patients in whom a vaginal component is more difficult. Although the majority of studies comparing conventional laparoscopy to robotic assisted laparoscopic surgery in the endometrial cancer space have been retrospective, small randomized prospective trials have been performed. Multiple retrospective trials suggest similar oncologic outcomes. Mäenpää and colleagues randomly assigned 101 endometrial cancer patients to conventional laparoscopy and robotic assisted laparoscopic surgery. The robotic approach was associated with a significant reduction in operative time, total time spent in the operative theater, and in conversions to laparotomy (Maenpaa, et al., 2016). Certainly, cost considerations must be accounted for given the large upfront capital costs associated with acquisition of robotic surgical equipment.

Increasing data suggest that even in grade I disease, as noted on endometrial biopsy, a significant number of patients have full surgical staging findings that would have an impact on further therapy. Ben-Shachar and colleagues, in evaluating 181 grade I endometrial cancers, found that 19% had grade change on hysterectomy specimen, 11% had extrauterine disease, 4% had lymph node metastasis, and 26% on final evaluation had high-risk intrauterine factors. Of note, the authors believed because of full surgical staging that 12% needed and received adjuvant therapy, and 17% who may have received postoperative treatment did not based on full surgical findings. Geisler and associates found in 349 patients that of those with grade I lesions, 16% had positive nodes and 3% had positive paraaortic nodes only. Of all positive nodes, 31% occurred in grade I lesions. As a result of these and other studies, it is advocated that all patients with endometrial cancer should have the benefit of surgical staging, which includes peritoneal cytology, TAH, and BSO, with SLN dissection. Obviously, complete evaluation of the entire peritoneal cavity and its contents should be performed, and any suspicious areas should be pathologically evaluated.

Radiation therapy

Radiation therapy remains a cornerstone of adjuvant therapy for endometrial cancer and has evolved considerably over time. Historically, nearly all patients received some form of preoperative or postoperative radiation therapy given the perception that risk for recurrence was high. Whereas GOG 33 defined relationships between depth of invasion and tumor grade and extrauterine spread after surgical staging, it also showed that most patients were not at risk for extrauterine disease spread. As such, the routine use of pelvic radiation therapy became increasingly questioned. To date, four large prospective randomized studies have compared external beam pelvic radiation with observation or vaginal brachytherapy in women with endometrial cancer ( Table 5.17 ). Aalders and colleagues compared VCB with or without pelvic radiation in 540 patients with early-stage endometrial cancer. Patients did not undergo lymph node dissection. The addition of pelvic radiation therapy reduced local failures (2% vs. 7%), but there was no difference in 5-year survival between groups (89% pelvic, 91% brachytherapy). Onsrud et al. updated the long-term follow-up for these patients. After a median 20.5 years of follow-up, no statistically significant difference was revealed in OS between treatment groups. However, women younger than age 60 years had significantly higher mortality rates after external-beam radiotherapy (EBRT) (HR, 1.36; 95% CI, 1.06 to 1.76) than the control group. The risk of secondary cancer increased after EBRT, especially in women younger than age 60 years (HR, 2.02; 95% CI, 1.30 to 3.15). The authors concluded that there was no survival benefit of external pelvic radiation in early-stage endometrial carcinoma. In women younger than age 60 years, pelvic radiation decreased survival and increased the risk of secondary cancer. Thus, adjuvant EBRT should be used with caution, especially in women with long life expectancies.

TABLE 5.17
Radiation in Early-Stage Carcinoma of the Endometrium
Local Recurrence (%) Survival (%)
  • Aalder

    • Surgery + VCB ( n = 277)

  • 91

    • Surgery VCB + RT ( n = 263)

  • 89

  • Creutzberg (PORTEC)

    • Surgery ( n = 300)

  • 85

    • Surgery + RT ( n = 354)

  • 81

  • Keyes (GOG 99)

    • Surgery ( n = 202)

  • 4

  • 86

    • Surgery + RT ( n = 190)

  • 2

  • 92

  • ASTEC/NCIC CTG EN.5

    • Surgery c ( n = 454)

  • 84

    • Surgery + RT ( n = 452)

  • 84

ASTEC , A Study in the Treatment of Endometrial Cancer; GOG, Gynecologic Oncology Group; NCIC CTG EN.5 , National Cancer Institute of Canada Clinical Trials Group Endometrial Cancer 5 trial; PORTEC, Post-Operative Radiation Therapy in Endometrial Cancer; RT, pelvic radiation therapy; VCB, vaginal cuff brachytherapy.

a P < .01.

b P < .001.

c 50% received vaginal brachytherapy.

d P = .038.

In the Dutch PORTEC trial, Creutzberg et al. enrolled 714 patients with grade I lesions with greater than 50% myometrial invasion, grade II lesions with any amount of invasion, or grade III with less than 50% invasion. Patients were randomly assigned postoperatively to receive either external radiation or observation. None of the patients were surgically staged, and all histologies were eligible. In the 654 eligible for follow-up, local and regional recurrences were less frequent in the radiation therapy group (4% vs. 14%) than in the observational group, but the 5-year survival rates were similar, 81% versus 85%, respectively. As published by Creutzberg et al., after 15 years, there remained no survival advantage and a trend for long-term risk of second cancers.

The GOG performed a phase III trial, GOG protocol 99, of surgery with or without adjuvant external beam pelvic radiation in intermediate-risk endometrial ACs. These included all women with any degree of myometrial invasion, any grade, and no evidence of lymph node metastasis (stage IB, IC, IIA occult, and IIB occult). All patients were required to have surgical staging with histologic evaluation of the lymph nodes. A total of 202 patients received no radiation (no adjuvant therapy [NAT]), and 190 received pelvic radiation. The median follow-up period was 69 months. The reported recurrence rate was 15% in the NAT group compared with 6% to 8% in the radiation group ( P = .007). Local recurrences were 9% versus 2%, respectively. The OS at 48 months was 86% for NAT and 92% for radiation, with intercurrent diseases accounting for half or more of the deaths in both groups. Deaths from disease were 8% in both the NAT versus radiation group. Grade III and IV toxicity by treatment was 5% versus 14% in the NAT and radiation, groups, respectively. Twelve of the 13 women who had isolated vaginal recurrences, and were initially randomized to the NAT arm were treated with radiation, and five have died of disease.

Importantly, GOG 99 allowed for the identification of a H-IR subgroup, in a post-hoc manner, that appeared to derive the greatest benefit from pelvic radiation. In the H-IR cohort, the recurrence rate was 27% in the observation arm, versus 13% after pelvic radiation. All other eligible patients were considered low–intermediate risk and had recurrence rates less than 1% to 3% regardless of the use of radiation therapy. H-IR was defined using a combination of age, grade, presence of LVSI, and depth of invasion.

Data from a pooled trial of the Medical Research Council (MRC), ASTEC and National Cancer Institute of Canada Clinical Trials Group Endometrial Cancer 5 (NCIC CTG (EN.5) randomized trials also showed no survival advantage for adjuvant EBRT in the treatment of endometrial cancer (5-year survival rate ~84% both groups). In this study including 906 patients randomly assigned to pelvic radiation or observation, 30% of patients underwent lymphadenectomy, and 50% received vaginal brachytherapy. Pelvic radiation reduced local failures (6% with observation vs. 3% after pelvic radiation). In a subsequent Cochrane review on the use of adjuvant radiation therapy for stage I endometrial cancer, the authors concluded that there is no survival advantage with routine use of pelvic radiation therapy and that benefits must be balanced against the toxicity and morbidity of therapy.

Essentially all studies evaluating pelvic radiation therapy show improved local control rates compared to observation. The local control, however, is largely a result of reduction in vaginal cuff recurrences. A considerable amount of data has been collected to evaluate vaginal recurrence and survival rate with surgery alone or combined therapy when mainly preoperative application of brachytherapy and surgery were used. Data have also been evaluated concerning the grade of the tumor ( Table 5.18 ) and, in some instances, the depth of myometrial involvement ( Table 5.19 ). In patients who had postoperative radiation, there appeared to be a lower incidence of vaginal vault recurrences, although there does not appear to be much difference in the grade I and grade II lesions. Vaginal vault recurrence did not appear to affect survival.

TABLE 5.18
Survival Rate in Stage I Carcinoma of the Endometrium With Regard to Grade and Treatment
From Pettersson F, editor: Annual report on the results of treatment in gynecological cancer, vol 21, Stockholm, 1991, International Federation of Gynecology and Obstetrics.
Grade SURVIVAL
Surgery Only (%) Combined Therapy (%)
I 1295/1375 (94) 2284/2389 (96)
II 488/510 (96) 1490/1721 (87)
III 100/135 (74) 398/498 (80)

TABLE 5.19
Recurrences in Stage I Carcinoma of the Endometrium With Regard to Depth of Invasion and Treatment
Modified from DiSaia PJ, Creasman WT, Boronow RC, et al: Risk factors in recurrent patterns in stage I endometrial carcinoma, Am J Obstet Gynecol 151:1009, 1985.
Recurrence Surgery and Radium Surgery and External Radiation
Endometrium only 6/88 (7%) 0/4 (0%)
Inner and mid thirds 3/68 (4%) 9/29 (31%)
Outer third 3/9 (33%) 11/24 (46%)

Results of a randomized prospective trial comparing adjuvant EBRT versus vaginal brachytherapy in 427 patients with intermediate-risk early-stage endometrial cancer have been reported (PORTEC-2). No differences in recurrence rate (vaginal failure 2% with pelvic radiation, 0.9% with vaginal brachytherapy) or OS (3-year survival rate ~90% both groups) were observed between the two treatment arms, suggesting that patients with intermediate-risk early-stage endometrial cancer can be treated with adjuvant vaginal brachytherapy alone. Although these findings have led many clinicians in the United States to move away from the routine use of EBRT in early-stage endometrial cancer, it is important to note that in the patients who did not undergo a lymph node dissection, pelvic failure rates were higher in the vaginal brachytherapy group (3.5% vs. 0.7%), and the study excluded higher-risk patients such as those with grade III or deeply invasive tumors. An additional study comparing pelvic radiation followed by VCB to VCB alone for “medium-risk” endometrial cancer by Sorbe et al. showed results similar to PORTEC-2, demonstrating that pelvic radiation reduced locoregional relapse rates (1% vs. 5%, P = .013) with no difference in OS. Sorbe et al. also reported a randomized trial of vaginal brachytherapy versus no further treatment in patients with low-risk endometrial cancer (grade I or II endometrioid cancer with <50% invasion). There was no significant reduction in vaginal recurrence in the group receiving brachytherapy (3% vs. 1%).

GOG protocol 249 was designed to determine whether adjuvant chemotherapy could improve cancer outcomes in patients with H-IR endometrial cancer ( ). This was based on the observation that both PORTEC-1 and GOG protocol 99 reported distant failure rates of 20% to 30% in high-risk patients. Eligible patients had International Federation of Gynecology and Obstetrics (2009) stage I endometrioid histology with H-IR criteria, stage II disease, or stage I to II serous or clear cell tumors. A total of 601 patients were randomly assigned between RT (45 to 50.4 Gy over 5 weeks) or VCB followed by intravenous paclitaxel 175 mg/m 2 (3 hours) plus carboplatin (area under the curve [AUC], 6) every 21 days for three cycles ( ). With a median follow-up of 53 months, the 60-month RFS was 0.76 for RT and 0.76 for brachytherapy followed by chemotherapy (HR 0.92; 90% CI 0.69 to 1.23). Furthermore, no difference in 60-month OS was identified between study arms (HR 1.04; 90% CI 0.71 to 1.52). Pelvic or paraaortic nodal recurrences were more common in the chemotherapy arm, 9% versus 4%. Ironically, patients with serous cancer accounted for 15% of patients enrolled on GOG 249, and although the expectation was that chemotherapy may benefit patients with this high-risk histology, there was no significant difference in PFS or OS when chemotherapy was combined with VCB.

Two additional pivotal clinical trials looked to better define the utility of radiation and chemotherapy in the management of locally advanced endometrial cancer. GOG protocol 258 was developed in an effort to address local recurrences, reported to occur in up to 20% of patients treated on the chemotherapy arm of GOG 122 ( ). The experimental arm was supported by prior studies, including RTOG-9708, which examined adjuvant cisplatin 50 mg/m 2 intravenously on days 1 and 29 plus volume-directed radiation therapy (45 Gray ± brachytherapy) followed by paclitaxel 175 mg/m 2 and carboplatin AUC 5 every 21 days for four cycles. GOG 258 enrolled a total of 813 patients with stage III or IVa endometrioid endometrial cancer, as well as stage I or II serous or clear cell carcinoma with positive washings. Patients were randomized to combined chemoradiation which consisted of cisplatin 50 mg/m 2 on days 1 and 29 together with volume-directed external beam radiation therapy, followed by carboplatin AUC of 5 to 6 plus paclitaxel at a dose of 175 mg/m 2 every 21 days for four cycles, with GCSF support or a chemotherapy-only regimen consisting of carboplatin AUC of 6 plus paclitaxel at a dose of 175 mg/m 2 every 21 days for six cycles. In the chemoradiotherapy group, external beam radiation therapy was delivered to the pelvis with or without paraaortic fields. The planned total dose was 45 Gy in 25 fractions at 180 cGy per fraction. Intensity modulated radiotherapy and vaginal brachytherapy were allowed only in the chemoradiotherapy group ( ). Nearly all enrolled patients (98%) had no gross residual disease, and 94% underwent some degree of lymph node assessment. At 60 months, the Kaplan–Meier estimate of the percentage of patients alive and relapse free was 59% (95% CI, 53 to 65) in the chemoradiotherapy group and 58% (95% CI, 53 to 64) in the chemotherapy only group (HR 0.90; 90% CI, 0.74 to 1.10). Chemoradiotherapy was associated with a lower 5-year incidence of vaginal recurrence (2% vs. 7%; HR 0.36; 95% CI, 0.16 to 0.82) and pelvic and paraaortic lymph node recurrence (11% vs. 20%; HR 0.43; 95% CI, 0.28 to 0.66) than chemotherapy alone, but distant recurrence was more common in association with chemoradiotherapy (27% vs. 21%; HR 1.36; 95% CI, 1.00 to 1.86). Furthermore, assessment of quality-of-life endpoints suggested both acute and long-term toxic effects of combination chemoradiation therapy, suggesting that chemotherapy alone was the most appropriate adjuvant regimen in this patient population.

In an analogous manner, PORTEC-3 was designed to compare adjuvant chemotherapy during and after radiotherapy (chemoradiotherapy) versus pelvic radiation alone in women with high-risk endometrial cancer. Eligible women had stage I, grade III endometrioid carcinoma with deep myometrial invasion or lymph-vascular space invasion (or both), stage II or III endometrioid AC, or stage I to III with serous or clear cell histology. Nearly half of the women enrolled on this trial had stage I or II disease. Patients were randomly assigned (1:1) to receive radiotherapy alone (48.6 Gy in 1.8 Gy fractions given on 5 days per week) or radiotherapy and chemotherapy (consisting of two cycles of cisplatin 50 mg/m 2 given during radiotherapy, followed by four cycles of carboplatin AUC5 and paclitaxel 175 mg/m 2 ) ( ). A total of 686 women were enrolled, with investigators showing an improvement in 5-year failure free survival in the chemoradiotherapy arm (HR 0.71, 95% CI 0.53 to 0.95; P = .022), but no significant improvement in OS (HR 0.76, 95% CI 0.54 to 1.06; P = .11) ( ). Grade 3 or worse adverse events were significantly more common in the chemoradiotherapy arm. When specifically examining patients with stage III disease, the investigators showed a more pronounced FFS advantage (HR 0.66, 95% CI 0.45 to 0.97) with a non-significant trend toward improved OS (HR 0.71, 95% CI 0.45 to 1.11).

An updated post-hoc survival analysis of PORTEC-3, completed at a median follow-up of nearly 73 months suggested both a significant OS and failure-free survival advantage with chemoradiotherapy ( ). This advantage appeared to be most pronounced in patients with stage III disease, as well as those with serous histology. Taken together, the results of GOG 258 as well as PORTEC-3 highlight the importance of chemotherapy in the management of patients with stage III endometrial carcinoma. Conversely, the results of GOG 249 and PORTEC-3 suggest that the addition of chemotherapy to radiation in patients with stage I or II disease may confer limited benefit with additional treatment-related toxicity.

Chemotherapy

The role of chemotherapy in the treatment of endometrial cancer continues to evolve. Traditionally reserved for patients with recurrent or metastatic disease, chemotherapy has become increasingly used in the first-line management of patients with advanced-stage and high-risk early-stage disease. Historically, radiation therapy has been the adjuvant of choice for patients at risk of recurrence. Although the overall prognosis of patients in adjuvant radiation trials has been favorable, approximately 20% of patients have a recurrence at distant sites, demonstrating the need for effective systemic therapy, as previously discussed.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here