Other rare uterine cancers: neuroendocrine tumors, yolk sac tumors, choriocarcinoma


Neuroendocrine tumors

Epidemiology

Incidence and mortality

Neuroendocrine carcinomas (NEC) of the endometrium, which includes both small-cell and large-cell variants, are rare, highly aggressive tumors that account for approximately 0.8% of endometrial carcinomas. They generally present with advanced stage disease. The literature consists of only case reports and small case series. In one case series, 32% of patients presented with early (stages I–II) disease, whereas 68% presented with advanced stage disease, with a predominance of incidence in the sixth and seventh decades of life. Another case series reports 26%, 5%, 36%, and 33% of patients present with stages I, II, III, and IV, respectively.

Median survival has been reported as 17 months with a 5-year survival of 38.3%. Stage for stage, 5-year survival for uterine neuroendocrine tumors (NETs) is significantly worse than survival for women with endometrioid adenocarcinoma of the uterus ( Table 13.1 ). Mean survival ranges from 12 months for advanced stage disease to 22 months for early stage disease. In a cohort of 25 patients, seven attained 5-year survival, of which three patients had stage I disease and four had stage III disease.

Table 13.1
Five-year survival rate by FIGO stage for uterine neuroendocrine carcinoma vs. endometrioid adenocarcinoma.
Adopted from Schlechtweg et al., GYN Onc, 2019.
Stage Neuroendocrine carcinoma Endometrioid carcinoma
IA 76.4% 85.4%
IB 65.0% 72.7%
II 37.5% 67.1%
IIIA 16.1% 58.2%
IIIB 45.0% 46.4%
IIIC 34.2% 53.1%
IVA 0.0% 28.8%
IVB 15.4% 27.7%

Etiology and risk factors

NEC of the endometrium are a histologically distinct subtype of endometrial carcinoma with a propensity of metastatic infiltration and an unfavorable prognosis. They tend to form bulky, intraluminal masses with deep myometrial involvement. They are subdivided into small- and large-cell tumors according to nuclear size. Endometrial NEC are often combined with other epithelial neoplasms, with 50%–80% of cases admixed with FIGO grade 1 or 2 endometrioid carcinoma.

Based on the largest population-based study ( N = 364) from the National Cancer Database (NCDB), compared to women with poorly differentiated endometrial cancer, women with NEC were more often black (13.7% neuroendocrine vs. 10.7% poorly differentiated endometrial cancer) and Hispanic (8.0% neuroendocrine vs. 5.3% poorly differentiated endometrial cancer). Additionally, as with other endometrial carcinomas, postmenopausal status is a known risk factor.

Pathology

Gross description

Grossly the tumors are large, endometrial based and typically with deep myometrial invasion. Tumor size ranges from 0.8 to 12 cm with a median tumor size of 6.0 cm. No gross pathognomonic features have been described.

Microscopic description

Small-cell neuroendocrine carcinoma (SCNECa) resembles that of pulmonary small-cell carcinoma, with “salt and pepper chromatin” and scant cytoplasm with nuclear molding. Large-cell neuroendocrine carcinoma (LCNECa) typically have neuroendocrine growth pattern, such as organoid/nesting ( Fig. 13.1A ), trabecular or corded. LCNECa characteristically have polygonal cells with abundant cytoplasm and prominent nucleoli ( Fig. 13.1B ). The tumors frequently have central geographic necrosis, numerous mitotic figures and apoptotic bodies. Tumors may show pure SCNECa or LCNECa or may show mixed small and large-cell morphology. NEC can also be mixed with other histotypes, including endometrioid adenocarcinomas, serous carcinomas and carcinosarcomas.

Fig. 13.1, (A) Large-cell neuroendocrine carcinoma with infiltrating nested pattern and foci of central necrosis. (B) Large-cell neuroendocrine carcinoma with large polygonal cells, abundant cytoplasm and prominent nucleoli.

Immunohistochemistry

Epithelial markers such as a pankeratin cocktail should be positive, while rare tumors that are negative for pancytokeratin cocktail may have expression of cytokeratin 18. In some cases, several markers may be necessary to establish epithelial origin. NEC are positive for at least one neuroendocrine marker in at least 10% of the tumor cells, including synaptophysin, chromogranin, and CD56. It should be noted though that non-NECs, such as an endometrioid FIGO grade 3 or undifferentiated carcinoma may show staining for any one of the neuroendocrine markers, and that interpretation of IHC should be in the context of the overall morphology. In some cases where the morphology and or IHC are not characteristic, a diagnosis of neuroendocrine differentiation or features may be used by the pathologist. p16 can be positive in a patchy or diffuse pattern, with no evidence of infection with high-risk HPV.

Differential diagnosis

The differential diagnosis includes NEC from other primary sites, such as extension from the cervix or rarely metastases from other sites. In large tumors that involve the cervix and endometrium, establishing the primary site requires immunohistochemical staining, as the tumors are histologically similar. p16, which is usually a surrogate marker of HPV infection, can be diffusely expressed by non-HPV associated pathways and is not helpful in this distinction. Therefore, testing for HR-HPV is usually required and a negative result would favor endometrial primary in the correct clinical setting. High-grade carcinomas with a solid or nested growth pattern, such as FIGO grade 3 endometrioid carcinoma or serous carcinoma, may warrant consideration of a NEC. While neuroendocrine markers may be exhibited in these tumors, IHC is focal and less than 10% of tumor cells. Undifferentiated/de-differentiated carcinoma is also a consideration, usually lacking a nested architecture and with a diffuse, sheet-like pattern of growth. Like other NEC, focal staining for neuroendocrine markers may be seen but should be less than 10% of tumor cells.

Molecular findings

Approximately 40% of a series of endometrial NEC showed loss of expression of mismatch repair proteins, with the most common pattern being loss of MLH1/PMS2. Molecular analysis on a separate series shows that endometrial NEC also classify into The Cancer Genome Atlas (TCGA) groups, with POLE- mutated/ultramutated (7%), MSI/hypermuted (43%) and TP53 mutated/copy number high (14%). Some SCNECa may show similar molecular profiles to those expected in typical pulmonary SCNECa ( RB1 deletion and TP53 mutations).

Diagnosis and workup

The differential diagnosis for NEC is broad and includes undifferentiated/dedifferentiated endometrial carcinoma, carcinosarcoma, poorly differentiated endometrioid carcinoma, serous carcinoma with a solid growth pattern and massive necrosis, primitive neuroectodermal tumors (PNETs), and carcinoid tumor. Additionally, NEC is often associated with tumors of other histologic types, including complex atypical hyperplasia, endometrioid carcinoma, carcinosarcoma, or serous carcinoma.

In a 25 patient series, the most common presenting symptom was vaginal bleeding (15 of 25 patients) with other less common presenting symptoms including abdominal pain, symptoms related to metastasis, and abnormal Pap test. Additional common presenting symptoms include postmenopausal bleeding and pelvic pain. Of note, in very rare cases, NEC can also present with signs of paraneoplastic disease and/or Cushing’s syndrome.

Workup begins with a complete physical exam including pelvic and rectovaginal examinations ( Table 13.2 ). The most common physical exam findings include abnormal vaginal bleeding, abdominal pain, and an enlarged uterus and/or a palpable pelvic mass. There is not a typical tumor marker that is elevated in NEC. CA-125 may be elevated in the setting of a mixed tumor, but this marker is not specific to NETs.

Table 13.2
Basic workup and testing for neuroendocrine carcinoma of the endometrium.
Component of workup Specific tests/considerations
Physical exam Pelvic and abdominal exams
Tumor markers Consider CA-125
Imaging
  • TVUS, CXR

  • CT C/A/P if high-grade carcinoma

  • Consider MRI A/P or PET

Diagnostic tests Endometrial biopsy or dilation and curettage
CXR : chest radiograph; CT C/A/P : computed tomography of chest/abdomen/pelvis; MRI A/P : magnetic resonance imaging of abdomen/pelvis; PET : positron emission tomography; TVUS : transvaginal ultrasound.

Data on the optimal pretreatment imaging are limited. Consideration can be given to obtaining a transvaginal pelvic ultrasound and a chest radiograph. If there is an abnormality on chest radiograph, a chest computed tomography (CT) can be obtained to further evaluate. If there is concern for high-grade carcinoma (e.g., if already diagnosed as high-grade tumor by an endometrial biopsy (EMB)), it is reasonable to obtain a CT of the chest, abdomen, and pelvis to evaluate for metastatic disease. Other initial imaging may be obtained based on symptoms and/or clinical concern for specific metastatic sites.

In some cases, an MRI abdomen and pelvis may be obtained. MRI findings are similar to other poorly differentiated carcinomas, with an ill-defined endometrial-myometrial border and heterogeneity of the mass indicating necrosis and hemorrhage. Consideration can also be given to positron emission tomography (PET).

Similar to other endometrial cancers, the gold standard for initial diagnosis is by pathologic diagnosis obtained from either an EMB or endometrial curettings obtained from a dilation and curettage (D&C).

Staging system

Uterine NEC is surgically staged utilizing the 2017 International Federation of Gynecology and Obstetrics (FIGO)/ Tumor, Node, Metastasis (TNM) system used for endometrial carcinoma ( Table 13.3 ).

Table 13.3
Staging for neuroendocrine carcinoma of the endometrium.
Stage Description
I Tumor confined to uterus
IA: < 50% myometrial invasion
IB: > 50% myometrial invasion
II Involvement of cervical stroma
III Involvement of serosa, adnexa, vagina, parametrium, or locoregional lymph nodes
IIIA: serosa and/or adnexa
IIIB: vagina or parametrium
IIIC: locoregional lymph nodes
IIIC1: pelvic lymph nodes
IIIC2: para-aortic lymph nodes
IV Involvement of bladder/bowel mucosa, or distant metastases
IVA: bladder/bowel mucosa
IVB: distant metastases

Prognostic factors

Factors associated with death in a NCDB series include: diagnosis at 80 years of age or older, stage (diagnosis at stage II or higher compared to stage I), and not receiving adjuvant chemotherapy. Women over the age of 80 have a hazard ratio for death of 2.4 compared to women less than 50 years old and patients who receive chemotherapy have a hazard ratio for death of 0.36 compared to those who do not get chemotherapy. In a smaller study of 42 patients, performance status, FIGO stage, surgery, and histologic subtype (pure type with worse prognosis than mixed type) were significant prognostic factors; in a multivariate analysis, only surgery and histologic subtypes remained significant. There was also a better prognosis in complete surgery cases (versus no surgery or incomplete surgery).

Prognostic factors for small-cell NETs in general (not specific to endometrial origin) include early detection and prompt surgical and adjuvant therapy.

Treatment of primary disease

Standard treatment guidelines do not exist given the rarity of NEC and lack of prospective data to guide therapy. Treatment regimens are largely based on traditional treatments of endometrial cancer and small-cell lung cancer. The majority of reported treatment regimens in the literature include surgical resection and adjuvant therapy with platinum-based chemotherapy with or without radiation therapy. Treatment usually begins with surgery for women without metastatic disease ( Fig. 13.2 ). Surgery generally includes hysterectomy, bilateral salpingo-oophorectomy (BSO), and lymph node assessment. For younger women, fertility preserving surgery can be considered on an individual basis but most patients should undergo BSO.

Fig. 13.2, Treatment of newly diagnosed uterine neuroendocrine carcinoma. BSO : bilateral salpingo-oophorectomy.

Adjuvant treatment is reasonable to consider given aggressive nature of this cancer type with high risk of recurrence and metastasis. In the largest reported series of cases, all patients underwent surgery, 60% received chemotherapy, and 28% received radiation. Compared to women with poorly differentiated endometrial carcinoma, those with NEC were more likely to receive adjuvant chemotherapy, likely owing to the tendency to present with later stage disease. In another series, all patients underwent surgery and 80% received adjuvant therapy. All patients undergoing surgery with the majority also receiving adjuvant chemotherapy is consistent with smaller series and case studies.

The most common reported chemotherapy regimens include cisplatin and etoposide. The following dosage regimen has been reported in the literature and is reasonable to consider for adjuvant treatment: cisplatin (80 mg/m 2 on day 1) and etoposide (80–120 mg/m 2 on days 1–3) over a 3-week interval. Of note, adjuvant treatment regimens used for NEC are largely based on data from small-cell lung cancer and neuroendocrine cervical cancer. A case series of SCNECa of the cervix demonstrated 83% recurrence-free survival (RFS) at 3 years with use of adjuvant etoposide and cisplatin. Indications for neoadjuvant chemotherapy are similar to those in other endometrial cancer types, and may be considered in the setting of advanced stage disease, though its use is rare in the reported literature.

In conclusion, there are no evidence-based therapeutic regimens for NEC due to its rarity, but surgery followed by adjuvant chemotherapy (most commonly cisplatin and etoposide) has been used with several patients obtaining long-term survival in case reports.

Surveillance for recurrence

Based on recommendations for surveillance of endometrial carcinoma, it is reasonable to consider physical exam every 3–6 months for 2–3 years and then every 6 months or annually with thorough review of systems and tumor markers (if initially elevated) at each visit, as well as imaging if clinically indicated. Evidence-based recommendations for surveillance are lacking.

Survival and patterns of failure

A large case series reports 5-year progression-free survival (PFS) and overall survival (OS). Five-year PFS is 81.8%, 50%, 32.5%, and 14.3% for FIGO stages I, II, III, and IV, respectively. OS is 88.9%, 100%, 46.7%, and 21.4% for FIGO stages I, II, III, and IV, respectively. Recurrence occurred in 50% of cases with a higher rate of hematogenous recurrence compared to lymphogenous or local recurrence.

Treatment of recurrent disease

There are no standard regimens due to rarity of the disease ( Table 13.4 ). One report in the literature describes the use of salvage chemotherapy with cisplatin (75 mg/m 2 ) and ifosfamide (5 g/m 2 ). Second-line therapies can be considered based on cervical and lung cancer data: cyclophosphamide/doxorubicin/vincristine (CAV), irinotecan/platinum (IP), or topotecan.

Table 13.4
Treatment of recurrence for neuroendocrine carcinoma of the endometrium.
Treatment options for recurrent disease Specific regimens/agents
Salvage chemotherapy Cisplatin and ifosfamide
Second-line therapies (used in cervical and lung cancer) Cyclophosphamide/doxorubicin/vincristine (CAV), irinotecan/platinum (IP), or topotecan
Targeted therapies PI3K and MEK inhibitors
Immunotherapy Nivolumab (PD-1 receptor inhibitor)

Based on preliminary data from NEC of the cervix, PI3K and MEK inhibitors are targeted therapies that may have therapeutic potential for treatment of endometrial NEC. One study looked at a newly established neuroendocrine cervical carcinoma cell line and found that when etoposide and cisplatin were combined with a PI3K inhibitor (which functions in the mTOR pathway), the growth of tumor cells was significantly reduced. Another study examined a MEK inhibitor, trametinib, which functions in the RAS pathway and is FDA-approved for treatment of unresectable or metastatic melanoma in patients with BRAF mutations, in the treatment of a patient with recurrent cervical neuroendocrine carcinoma with a KRAS mutation; she has been on trametinib for 8 cycles and is without evidence of disease. Based on data from pulmonary small-cell tumors, a number of other targeted therapies have been studied including angiogenesis inhibitors, mTOR inhibitors, and PARP inhibitors.

There is a case report evaluating the use of a programmed cell death 1 (PD-1) receptor inhibitor, nivolumab, in the treatment of a patient with recurrent, metastatic SCNECa of the cervix. The patient experienced a complete response and remained free of disease 4 months after stopping treatment. Further research is needed to see if immunotherapy is a reasonable approach to recurrent NEC of the cervix and/or endometrium.

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