1540 1413 Article p181

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 29

NCCN CLINICAL PRACTICE GUIDELINES IN ONCOLOGY

Uterine Neoplasms, Version 1.2023


Nadeem Abu-Rustum, MD1,*; Catheryn Yashar, MD2,*; Rebecca Arend, MD3; Emma Barber, MD4; Kristin Bradley, MD5;
Rebecca Brooks, MD6; Susana M. Campos, MD, MPH, MS7; Junzo Chino, MD8; Hye Sook Chon, MD9; Christina Chu, MD10;
Marta Ann Crispens, MD11; Shari Damast, MD12; Christine M. Fisher, MD, MPH13; Peter Frederick, MD14;
David K. Gaffney, MD, PhD15; Robert Giuntoli II, MD16; Ernest Han, MD, PhD17; Jordan Holmes, MD, MPH18;
Brooke E. Howitt, MD19; Jayanthi Lea, MD20; Andrea Mariani, MD21; David Mutch, MD22; Christa Nagel, MD23;
Larissa Nekhlyudov, MD, MPH7; Mirna Podoll, MD11; Ritu Salani, MD, MBA24; John Schorge, MD25; Jean Siedel, DO, MS26;
Rachel Sisodia, MD7; Pamela Soliman, MD, MPH27; Stefanie Ueda, MD28; Renata Urban, MD29;
Stephanie L. Wethington, MD, MSc30; Emily Wyse, BA31; Kristine Zanotti, MD32; Nicole R. McMillian, MS, CHES33;
and Shaili Aggarwal, PhD33

ABSTRACT NCCN CATEGORIES OF EVIDENCE AND CONSENSUS


Category 1: Based upon high-level evidence, there is uniform
Adenocarcinoma of the endometrium (also known as endometrial can- NCCN consensus that the intervention is appropriate.
cer, or more broadly as uterine cancer or carcinoma of the uterine cor- Category 2A: Based upon lower-level evidence, there is uniform
pus) is the most common malignancy of the female genital tract in the NCCN consensus that the intervention is appropriate.
United States. It is estimated that 65,950 new uterine cancer cases will
Category 2B: Based upon lower-level evidence, there is
have occurred in 2022, with 12,550 deaths resulting from the disease.
NCCN consensus that the intervention is appropriate.
Endometrial carcinoma includes pure endometrioid cancer and carcino-
mas with high-risk endometrial histology (including uterine serous carci- Category 3: Based upon any level of evidence, there is major
noma, clear cell carcinoma, carcinosarcoma [also known as malignant NCCN disagreement that the intervention is appropriate.
mixed M€ ullerian tumor], and undifferentiated/dedifferentiated carci-
noma). Stromal or mesenchymal sarcomas are uncommon subtypes ac- All recommendations are category 2A unless otherwise noted.
counting for approximately 3% of all uterine cancers. This selection from
Clinical trials: NCCN believes that the best management of
the NCCN Guidelines for Uterine Neoplasms focuses on the diagnosis, any patient with cancer is in a clinical trial. Participation in
staging, and management of pure endometrioid carcinoma. The com- clinical trials is especially encouraged.
plete version of the NCCN Guidelines for Uterine Neoplasms is available
online at NCCN.org.
J Natl Compr Canc Netw 2023;21(2):181–209 PLEASE NOTE
doi: 10.6004/jnccn.2023.0006 The NCCN Clinical Practice Guidelines in Oncology (NCCN
Guidelines®) are a statement of evidence and consensus of the
authors regarding their views of currently accepted approaches
to treatment. Any clinician seeking to apply or consult the NCCN
Guidelines is expected to use independent medical judgment in
the context of individual clinical circumstances to determine any
1
Memorial Sloan Kettering Cancer Center; 2UC San Diego Moores Cancer patient’s care or treatment. The National Comprehensive Can-
Center; 3O’Neal Comprehensive Cancer Center at UAB; 4Robert H. Lurie cer Network® (NCCN®) makes no representations or warranties
Comprehensive Cancer Center of Northwestern University; 5University of of any kind regarding their content, use, or application and dis-
Wisconsin Carbone Cancer Center; 6UC Davis Comprehensive Cancer Center; claims any responsibility for their application or use in any way.
7
Dana-Farber/Brigham and Women’s Cancer Center | Massachusetts General
Hospital Cancer Center; 8Duke Cancer Institute; 9Moffitt Cancer Center; The complete NCCN Guidelines for Uterine Neoplasms are
10
Fox Chase Cancer Center; 11Vanderbilt-Ingram Cancer Center; 12Yale Cancer not printed in this issue of JNCCN but can be accessed on-
Center/Smilow Cancer Hospital; 13University of Colorado Cancer Center; line at NCCN.org.
14
Roswell Park Comprehensive Cancer Center; 15Huntsman Cancer Institute at © 2023 National Comprehensive Cancer Network. All rights
the University of Utah; 16Abramson Cancer Center at the University of reserved. The NCCN Guidelines and the illustrations herein
Pennsylvania; 17City of Hope National Medical Center; 18Indiana University may not be reproduced in any form without the express writ-
Melvin and Bren Simon Comprehensive Cancer Center; 19Stanford Cancer ten permission of NCCN.
Institute; 20UT Southwestern Simmons Comprehensive Cancer Center;
21
Mayo Clinic Cancer Center; 22Siteman Cancer Center at Barnes-Jewish
Disclosures for the NCCN Uterine Neoplasms Panel
Hospital and Washington University School of Medicine; 23The Ohio State
University Comprehensive Cancer Center - James Cancer Hospital and Solove At the beginning of each NCCN Guidelines Panel meeting,
Research Institute; 24UCLA Jonsson Comprehensive Cancer Center; 25St. Jude panel members review all potential conflicts of interest.
Children’s Research Hospital/The University of Tennessee Health Science NCCN, in keeping with its commitment to public transparency,
Center; 26University of Michigan Rogel Cancer Center; 27The University of Texas publishes these disclosures for panel members, staff, and
MD Anderson Cancer Center; 28UCSF Helen Diller Family Comprehensive NCCN itself.
Cancer Center; 29Fred Hutchinson Cancer Center; 30The Sidney Kimmel Individual disclosures for the NCCN Uterine Neoplasms Panel
Comprehensive Cancer Center at Johns Hopkins; 31Patient advocate; members can be found on page 209. (The most recent version
32
Case Comprehensive Cancer Center/University Hospitals Seidman Cancer of these guidelines and accompanying disclosures are avail-
Center and Cleveland Clinic Taussig Cancer Institute; and 33National able at NCCN.org.)
Comprehensive Cancer Network.
The complete and most recent version of these guidelines is
*Discussion Writing Committee Member. available free of charge at NCCN.org.

JNCCN.org | Volume 21 Issue 2 | February 2023 181


NCCN GUIDELINES® Uterine Neoplasms, Version 1.2023

Overview grade disease.9 In addition to grade and depth of myome-


Endometrial carcinoma is a malignant epithelial tumor trial invasion, other risk factors associated with poor prog-
that forms in the inner lining, or endometrium, of the nosis include age, lymph node involvement, tumor size,
uterus. Risk factors for endometrial carcinomas include lymphovascular space invasion (LVSI), and lower uterine
increased levels of estrogen (caused by obesity, diabetes, segment invasion.10,11 Depth of myometrial invasion is
unopposed supplemental estrogen, and high-fat diet), early considered one of the critical criteria for evaluation of
age at menarche, nulliparity, late age at menopause, Lynch surgical-pathologic staging.12,13 To further improve out-
syndrome, ages between 55 and 64 years, and tamoxifen comes for patients with this disease, physicians need to
use.1–6 Data show that almost 67% of patients with adeno- identify high-risk patients and to tailor treatment appro-
carcinoma of the endometrium are diagnosed with disease priately to provide the best long-term survival, including
confined to the uterus at diagnosis.7 Regional and distant quality of life. The panel suggests that gynecologic oncol-
disease comprise approximately 21% and 8% of cases, ogists be involved in the primary management of all
respectively. patients with endometrial cancer.
Many physicians believe that adenocarcinoma of the
endometrium is a more treatable malignancy because the Initial Evaluation
early symptoms of metrorrhagia or postmenopausal vaginal For patients with known or suspected uterine neoplasms,
bleeding often trigger patients to seek care when the dis- the initial preoperative evaluation/workup for known or
ease is at an early and treatable stage. However, data show suspected malignancy includes a history and physical ex-
that the mortality rate for uterine cancer has increased amination, complete blood count (including platelets), con-
more rapidly than the incidence rate.8 This increased mor- sideration of liver function tests/renal function tests or
tality may be related to an increased rate of advanced-stage chemistry profile, expert pathology review with additional
cancers, high-risk histologies (eg, serous carcinomas), and endometrial biopsy as indicated, imaging, recommendation
patients being diagnosed at an age $65 years. Analysis of of genetic evaluation of tumor and for inherited cancer risk,
SEER data suggests that survival is increased in patients and consideration of germline testing and/or multigene
who are younger, have early-stage disease, and have lower- panel testing (see “Initial Evaluation” and “Principles of

182 © JNCCN—Journal of the National Comprehensive Cancer Network | Volume 21 Issue 2 | February 2023
Uterine Neoplasms, Version 1.2023 NCCN GUIDELINES®

Imaging” in the NCCN Guidelines for Uterine Neoplasms, study identified 4 major clinically significant molecular
available at NCCN.org).14 Preoperative imaging and biopsy subtypes with differing clinical prognosis: POLE (DNA
may help to identify uterine sarcomas, although biopsy sen- polymerase epsilon) mutated, microsatellite instability-
sitivity is less than that for endometrial cancer. An expert high (MSI-H), copy number low, and copy number high
pathology review will determine whether a patient has (associated with abnormal p53 expression/TP53 mutation).
a malignant epithelial tumor or a stromal/malignant mes- The POLE-mutated tumors harbor POLE exonuclease
enchymal tumor. Given the typical age group at risk for domain mutations. The copy number high group is
uterine neoplasms (ie, $55 years) and the presence of characterized by frequent TP53 alterations. These geno-
comorbid illnesses, also see the NCCN Guidelines for Older mic classes are also associated with characteristic phe-
Adult Oncology (available at NCCN.org). notypes. The endometrial cancers with POLE mutations
may be high-grade tumors with deep myometrial inva-
Molecular Analysis and Genetic Factors sion and LVSI, and usually have a good prognosis.16,17
Most endometrial cancer (95%) is caused by sporadic The copy number high is the most aggressive subtype and
(somatic) mutations. However, genetic mutations cause requires multimodality treatment, which almost always in-
endometrial cancer in about 5% of patients, which oc- cludes chemotherapy. The MSI-H tumors have an inter-
curs 10 to 20 years before sporadic cancer.15 Since there mediate prognosis but could be associated with other
is increasing overlap in histopathologic features of these genetic cancer predisposition, and sensitivity to chemo-
tumors, molecular analysis (eg, identification of charac- therapy and immune checkpoint inhibition has been un-
teristic translocations and/or mutations) and subtype der investigation. Further studies have attempted to study
classification are useful in selecting appropriate thera- the association of TCGA subgroups with histologic features
pies. The Cancer Genome Atlas (TCGA) study performed such as tumor grade and histologic type.18
an integrated genomic, transcriptomic, and proteomic The NCCN Guidelines for Uterine Neoplasms include
analysis of 373 endometrial carcinomas including low- a diagnostic algorithm for integrated genomic-pathologic
grade endometrioid, high-grade endometrioid, and se- classification of endometrial carcinomas based on the
rous carcinomas for their molecular classification. The TCGA study and add that the decision to use molecular

JNCCN.org | Volume 21 Issue 2 | February 2023 183


NCCN GUIDELINES® Uterine Neoplasms, Version 1.2023

testing/classification depends on resource availability and than a germline mutation.22 Genetic counseling, molecular
each center’s multidisciplinary team. The panel encourages analysis, and testing are recommended for patients with
comprehensive genomic profiling via a validated and/or all other MMR deficiencies. Patients with a significant
FDA-approved assay in the initial evaluation of uterine neo- family history of endometrial and/or colorectal cancer
plasms to help facilitate cancer diagnoses. The panel also (even for those without MMR defects, who are MSI-stable,
encourages ancillary studies of POLE mutation status, mis- or those without screening) should be referred for ge-
match repair (MMR)/MSI, and aberrant p53 expression to netic counseling and evaluation (See “Lynch Syndrome
complement the morphologic assessment of histologic tu- [Hereditary Non-Polyposis Colorectal Cancer]” in the
mor type. In addition, the panel includes consideration for NCCN Guidelines for Genetic/Familial High-Risk Assess-
NTRK gene fusion testing for metastatic or recurrent endo- ment: Colorectal, available at NCCN.org). Screening for
metrial carcinoma, and for tumor mutational burden (TMB) genetic mutations should be considered, especially for
testing through a validated and/or FDA-approved assay. patients ,50 years of age.6,15,19,26 If these patients have
Screening of the tumor for defective DNA MMR using Lynch syndrome, they are at a higher lifetime risk (#60%)
immunohistochemistry (IHC) and/or MSI is used to iden- for endometrial cancer; thus, close monitoring and dis-
tify which patients should undergo mutation testing for cussion of risk-reducing strategies is recommended.4,19,27
Lynch syndrome (see “Lynch Syndrome” in the NCCN In addition, their relatives may have Lynch syndrome. For
Guidelines for Colorectal Cancer Screening; available at patients and family members with Lynch syndrome but
NCCN.org).19–25 At a minimum, universal testing of endo- without endometrial cancer, a yearly endometrial biopsy
metrial tumors for defects in DNA MMR is recommended is recommended to assess for cancer.28,29 This strategy
(eg, MLH1, MSH2, MSH6, PMS2). MSI testing is recom- also enables select patients to defer surgery (and surgical
mended if MMR results are equivocal. Testing may be per- menopause) and to preserve fertility. Prophylactic hyster-
formed on the initial biopsy or dilation and curettage ectomy/bilateral salpingo-oophorectomy (BSO) is recom-
(D&C) material or the final hysterectomy specimen. MLH1 mended after childbearing is complete.30,31 In addition,
loss should be further evaluated for MLH1 promoter meth- interventions to decrease the risk from colorectal cancer
ylation to assess for a somatic epigenetic process rather are recommended (eg, annual colonoscopy).

184 © JNCCN—Journal of the National Comprehensive Cancer Network | Volume 21 Issue 2 | February 2023
Uterine Neoplasms, Version 1.2023 NCCN GUIDELINES®

Diagnosis and Workup and/or FDG-PET/CT may be used to assess disease ex-
Currently, no validated screening test for endometrial car- tent and to evaluate for metastatic disease as indicated
cinoma exists.32,33 About 90% of patients with endometrial based on clinical symptoms, physical findings, or abnor-
carcinoma have metrorrhagia, most commonly in the mal laboratory findings.37–40 In patients with extrauterine
postmenopausal period. Diagnosis can usually be made disease, a serum CA-125 assay may be helpful in moni-
by an office endometrial biopsy, with a false-negative rate toring clinical response. However, serum CA-125 levels
of about 10%.34,35 Thus, a negative endometrial biopsy in can be falsely increased in patients who have peritoneal
a symptomatic patient must be followed by a fractional inflammation/infection or radiation injury, may be nor-
D&C under anesthesia.34,36 Hysteroscopy may be helpful mal in patients with isolated vaginal metastases, and
in evaluating the endometrium for lesions, such as a may not predict recurrence in the absence of other clini-
polyp, if the patient has persistent or recurrent bleeding. cal findings.41,42
Endometrial biopsy may not be accurate for diagnosing
malignancies of the uterine wall such as mesenchymal tu- Disease Staging
mors. The histologic information from the endometrial The FIGO system is most commonly used for staging
biopsy (with or without endocervical curettage) is suffi- uterine cancer. The original 1970 criteria for staging en-
cient for planning definitive treatment. dometrial cancer only used information gained from the
presurgical evaluation (including physical examination
Imaging and Laboratory and diagnostic fractional D&C). The 1970 staging system
For detailed imaging recommendations by stage and is rarely used today (eg, reserved for when the patient is
planned treatment approach, see “Principles of Imag- not a surgical candidate).
ing” in the NCCN Guidelines for Uterine Neoplasms Several studies demonstrated that clinical staging was
(available at NCCN.org). Consideration of preoperative inaccurate and did not reflect actual disease extent in 15%
chest imaging (chest X-ray) is recommended. Based on to 20% of patients.43 This reported understaging and,
the fertility-sparing or non–fertility-sparing treatment more importantly, the ability to identify multiple prognos-
criteria, other imaging tests such as CT, MRI, ultrasound, tic factors with a full pathologic review made possible with

JNCCN.org | Volume 21 Issue 2 | February 2023 185


NCCN GUIDELINES® Uterine Neoplasms, Version 1.2023

surgical staging motivated a change in the staging classifica- patients with positive para-aortic nodes.46 To maintain
tion. Therefore, in 1988, FIGO modified its staging system to consistency, the NCCN panel has reinterpreted historical
emphasize thorough surgical/pathologic assessment of studies using the 1988 FIGO staging system to reconcile
data, such as histologic grade, myometrial invasion, and the those studies with the 2009 staging system.
extent and location of extrauterine spread (including retro- In the 2009 FIGO staging, the presence of positive peri-
peritoneal lymph node metastases).44 FIGO updated and re- toneal cytology no longer increases the disease stage, as its
fined the surgical/pathologic staging criteria for uterine importance as an independent risk factor has been called
neoplasms in 2009.45–47 Separate staging systems for malig- into question.47 However, FIGO and AJCC continue to rec-
nant epithelial tumors and uterine sarcomas are now avail- ommend that peritoneal washings be obtained and results
able (see “Staging” section of the algorithm at NCCN.org). In be recorded (see “Principles of Evaluation and Surgical
2017, the AJCC Cancer Staging Manual was further updated Staging,” next section).49
(which took effect in January 2018).48
The 2009 FIGO staging system streamlined stages I Principles of Evaluation and Surgical Staging
and II of endometrial carcinoma. These revisions were for Endometrial Carcinoma
made because the survival rates for some of the previous Staging should be done by a team with expertise in imag-
substages were similar.46 Currently stage IA describes ing, pathologic evaluation, and surgery. The amount of
tumors with ,50% myometrial invasion, and stage IB surgical staging that is necessary to determine disease sta-
describes those with $50% myometrial invasion. Stage II tus depends on preoperative and intraoperative assessment
describes patients with tumors that invade the cervical by experienced surgeons. Pathologic nodal assessment for
stroma. Patients with uterine-confined disease and endocer- apparent uterine-confined endometrial cancer informs
vical glandular involvement (mucosal involvement) without both stage and adjuvant therapy. However, if final pathol-
cervical stromal invasion are no longer considered stage II.46 ogy shows a noninvasive endometrioid histology, nodal as-
Stage IIIC is subdivided into IIIC1 (pelvic nodal involvement sessment can be eliminated. The NCCN sentinel lymph
alone) and IIIC2 (para-aortic involvement 6 pelvic node node (SLN) algorithm is recommended if sentinel node
involvement), reflecting the inferior survival in those mapping is used.

186 © JNCCN—Journal of the National Comprehensive Cancer Network | Volume 21 Issue 2 | February 2023
Uterine Neoplasms, Version 1.2023 NCCN GUIDELINES®

Pathology should undergo ultrastaging for the detection of low-


An expert pathologic review determines the specific volume metastases. Ultrastaging commonly entails thin
histotype of the tumor (endometrioid, serous, clear cell, serial sectioning of the gross SLN and review of multiple
carcinosarcoma, or undifferentiated/dedifferentiated). H&E-stained sections with or without cytokeratin IHC
Morphologic evaluation of endometrial carcinoma to for all blocks of SLN. There is no standard protocol for
determine histologic type—especially in high-grade lymph node ultrastaging. See “Principles of Pathology”
cancers—is challenging, and issues exist regarding in the NCCN Guidelines for Endometrial Carcinoma
diagnostic reproducibility. The pathologic assessment (available at NCCN.org). The Protocol for the Examina-
of the uterus and the nodes is described in the algo- tion of Specimens From Patients With Carcinoma and
rithm. The assessment of the uterus includes the hys- Carcinosarcoma of the Endometrium from the College
terectomy type, specimen integrity, tumor site and size, of American Pathologists is a useful guide (available at
histologic type and grade if applicable, myometrial in- https://cap.objects.frb.io/protocols/cp-femalereproductive-
vasion (depth of invasion in mm/myometrial thickness endometrium-18protocol-4100.pdf). This protocol was re-
in mm), cervical stromal involvement, and LVSI. Pathol- vised to reflect the updated pTNM requirements from the
ogists may be asked to quantify LVSI. The current defi- AJCC Cancer Staging Manual (8th edition) and 2015 FIGO
nition of substantial LVSI is $4 LVSI-involved vessels in Cancer Report.48,51
$1 hematoxylin and eosin (H&E) slide (for clinically rel- Estrogen receptor (ER) testing is recommended in
evant LVSI in endometrial cancer).50 the setting of stage III, IV, or recurrent endometrioid car-
The pathologic assessment should also include as- cinoma. Evaluation of HER2 overexpression should also
sessment of involvement by other tissues such as the fal- be considered. Rottmann et al52 recently showed that 16%
lopian tubes, ovaries, vagina, parametrium, peritoneum, of 80 gynecologic carcinosarcomas (including uterine car-
and omentum. The assessment of peritoneal/ascitic fluid cinosarcoma) showed HER2 overexpression and amplifi-
cytology should also be obtained. If nodal resection was cation when using the 2013 ASCO/College of American
performed, the level of nodal involvement (ie, pelvic, Pathologists scoring system. Similar results were reported
common iliac, para-aortic) should be determined. SLNs by several studies,53,54 including by Yoshida et al55 and

JNCCN.org | Volume 21 Issue 2 | February 2023 187


NCCN GUIDELINES® Uterine Neoplasms, Version 1.2023

Jenkins et al.56 The panel recommends HER2 IHC testing trial data support routine full lymphadenectomy,61 al-
(with reflex to HER2 fluorescence in situ hybridization for though some retrospective studies have suggested that
equivocal IHC) for possible treatment of advanced-stage it is beneficial.62 Two randomized clinical trials from
or recurrent serous endometrial carcinoma or carcinosar- Europe reported that routine lymph node dissection did
coma. HER2 IHC testing should also be considered in not improve the outcome of patients with endometrial
TP53-mutated/p53 abnormal endometrial carcinoma re- cancer, but lymphadenectomy did identify those with
gardless of histotype. nodal disease.63,64 However, these findings remain a
As the grade of the tumor increases, the accuracy of point of contention.65–67 To avoid overinterpretation of
intraoperative evaluation of myometrial invasion decreases these results, it is important to address the limitations of
(ie, assessment by gross examination of fresh tissue).57 In these randomized studies, including selection of patients,
one study, the depth of invasion was accurately deter- extent of lymph node dissection, and standardization of
mined by gross examinations in 87.3% of grade 1 lesions, postoperative therapy.68 One of the trials did not stan-
64.9% of grade 2 lesions, and 30.8% of grade 3 lesions.58 dardize adjuvant treatment after staging surgery with
Studies show that in 15% to 20% of cases, the preoperative lymphadenectomy and this has been identified as a
grade (as assessed by endometrial biopsy or curettage) is weakness of the trial and may have contributed to the
upgraded on final pathologic evaluation of the hysterec- lack of difference in recurrence and survival in the 2
tomy specimen.59 groups.62 The other concerns include the lack of central
pathology review, subspecialty of surgeons, and ade-
Lymphadenectomy quacy of statistical power.
Previously, a full standard lymphadenectomy (ie, dissec- Decisions about whether to perform lymphadenec-
tion and assessment of both pelvic and para-aortic no- tomy, and, if done, to what extent (eg, pelvic nodes only or
des) was recommended for all patients; however, to both pelvic and para-aortic nodes), can be made based on
decrease side effects, a more selective and tailored nodal preoperative and intraoperative findings. Criteria have
evaluation approach that includes the SLN algorithm is been suggested as indicative of low risk for nodal metasta-
recommended by the NCCN panel.60 No randomized ses: (1) ,50% myometrial invasion; (2) tumor ,2 cm; and

188 © JNCCN—Journal of the National Comprehensive Cancer Network | Volume 21 Issue 2 | February 2023
Uterine Neoplasms, Version 1.2023 NCCN GUIDELINES®

(3) well or moderately differentiated histology.69,70 However, panel recommends that nodal evaluation be performed in
this may be difficult to accurately determine before final pa- patients with endometrial carcinoma, including para-aortic
thology results are available. If expert gynecologic pathol- lymphadenectomy in high-risk patients (see “Principles of
ogy is available, a frozen section to assess myoinvasion can Evaluation and Surgical Staging,” page 186).5 SLN mapping
be obtained and lymphadenectomy avoided if no myoinva- is the preferred alternative to full lymphadenectomy in the
sion or cervical invasion is identified.71 setting of apparent uterine-confined disease. The SLN sur-
Nodal evaluation will identify those patients with nodal gical algorithm is described subsequently. Lymphadenec-
metastases. Identification of metastatic disease guides ap- tomy is not recommended for patients with uterine
propriate adjuvant treatment that has been shown to im- sarcoma because metastasis to the nodes is unusual.
prove survival and decrease locoregional recurrence.
The question of whether to add para-aortic lympha- SLN Mapping
denectomy to pelvic node dissection has been debated. The section on surgical staging (see “Principles of Evalua-
Prior studies have shown conflicting information regard- tion and Surgical Staging,” page 186) includes recommen-
ing the risk of para-aortic nodal metastases in patients dations about SLN mapping. SLN mapping may be
without disease in the pelvic nodes.43,69,72,73 Para-aortic considered for patients without suspicion of metastatic dis-
lymphadenectomy up to the renal vessels may be consid- ease by preoperative imaging and no obvious extrauterine
ered for selective patients, including those with pelvic disease at exploration.75–79 In SLN mapping, dye is injected
lymphadenectomy or high-risk histologic features. Many into the cervix, which travels to the sentinel nodes. This has
surgeons do not do a full lymphadenectomy in patients emerged as a useful and validated technique for identifica-
with grade 1 early-stage endometrial cancer.60 tion of lymph nodes that are at high risk for metastases (ie,
In summary, lymph node dissection identifies patients SLN in patients with early-stage endometrial cancer).80 Su-
requiring adjuvant treatment with radiation therapy (RT) perficial (1–3 mm) and optional deep (1–2 cm) cervical in-
and/or systemic therapy.74 A subset of patients may not jection leads to dye delivery to the main layers of lymphatic
benefit from lymphadenectomy; however, it may be diffi- channel origins in the cervix and corpus, namely the
cult to preoperatively identify these patients. The NCCN superficial subserosal, intermediate stromal, and deep

JNCCN.org | Volume 21 Issue 2 | February 2023 189


NCCN GUIDELINES® Uterine Neoplasms, Version 1.2023

submucosal lymphatic sites of origin.81 Injection into the confined disease (see Figure 4 in “Principles of Evaluation
uterine cervix provides excellent dye penetration to the and Surgical Staging,” page 188).75,87 SLN mapping may be
uterine vessels and main uterine lymphatic trunks that most appropriate for those at low to intermediate risk for
condense in the parametria and appear in the broad liga- metastases and/or for those who may not tolerate a stan-
ment leading to pelvic and occasionally para-aortic senti- dard lymphadenectomy.79,81,88–93 SLN identification should
nel nodes. The uterine body lymphatic trunks commonly always be done before hysterectomy, except in cases where a
cross over the obliterated umbilical artery with the most bulky uterus must be removed to allow access to iliac vessels
common location of pelvic SLN being medial to the exter- and lymph nodes. For example, suspicious or grossly en-
nal iliac, ventral to the hypogastric, or in the superior part larged nodes should be removed regardless of SLN mapping
of the obturator region. A less common location is usually results. In SLN mapping, the surgeon’s expertise and atten-
seen when the lymphatic trunks do not cross over the tion to technical detail are critical. Patients may be able to
obliterated umbilical and move cephalad following the avoid the morbidity of a standard lymphadenectomy with
mesoureter; in these cases, the SLN is usually seen in the SLN mapping.92,94 Because SLNs identify the primary lym-
common iliac presacral region (see Figures 1–3 in phatic pathway, this increases the yield of finding metastatic
“Principles of Evaluation and Surgical Staging,” page disease during the mapping process. For cases of failed SLN
187). The radiolabeled colloid most commonly injected mapping, reinjection of the cervix may be considered.
into the cervix is technetium-99m (99mTc); colored dyes However, if SLN mapping fails, a reflex side-specific
are available in a variety of forms (isosulfan blue, 1%; nodal dissection should be performed and any suspi-
methylene blue, 1%; and patent blue, 2.5% sodium). In- cious or grossly enlarged nodes should be removed re-
docyanine green recently emerged as a useful imaging gardless of mapping.75,93
dye that requires a near-infrared camera for localization, A literature review and consensus recommendations
provides a very high SLN detection rate, and is commonly for SLN mapping in endometrial cancer were released by
used in many practices at the present time.82–86 the Society of Gynecologic Oncology (SGO).79 Close ad-
A surgical SLN algorithm is proposed to decrease the herence to the NCCN SLN surgical algorithm was found to
false-negative rate in patients with apparent uterine- result in accurate prediction of pelvic lymph node

190 © JNCCN—Journal of the National Comprehensive Cancer Network | Volume 21 Issue 2 | February 2023
Uterine Neoplasms, Version 1.2023 NCCN GUIDELINES®

metastasis with a less than 5% false-negative rate.75 Addi- positive SLN underwent external beam RT (EBRT) and che-
tionally, results were published from the FIRES trial, which motherapy at a higher rate than those with a negative SLN.
compared SLN mapping to lymphadenectomy for endo- In patients with a detected SLN, recurrence-free survival
metrial cancer in the largest multicenter prospective study (RFS) at 50 months was 84.7%, and no difference was de-
to date (n5385).81 Mapping of at least one SLN was suc- tected between patients with and without a positive SLN
cessful in 86% of patients; sensitivity was 97.2% (95% CI, (P5.5).97
85.0–100), and negative predictive value was 99.6% (95% SLN mapping should be done in institutions with ex-
CI, 97.9–100). pertise in this procedure. If patients have apparent distant
A systematic review of 17 studies with n .30 patients metastatic disease (based on imaging and/or surgical explo-
revealed detection rates of 60% to 100%; detection rates for ration), removal of nodes for staging purposes is not neces-
studies with larger cohorts (n .100) were $80%. Retrospec- sary because it will not change management.40,98
tive application of a surgical algorithm generated 95% sensi- Historically, SLN mapping was controversial in pa-
tivity, 99% predictive value, and a 5% false-negative rate.95 tients with high-risk histology (eg, serous carcinoma, clear
Another systematic review and meta-analysis of 55 studies cell carcinoma, carcinosarcoma).60,99 However, SLN map-
with n .10 patients (n54915) generated an overall detec- ping in patients with high-risk histologies (ie, grade 3, se-
tion rate of 81% with a 50% bilateral pelvic node detection rous, clear cell, carcinosarcoma) has been reported with
rate and 17% para-aortic detection rate.71 In a retrospective promising results as a potential alternative to complete
analysis of patients with early-stage endometrial cancer lymphadenectomy.93,100,101 A recent multi-institutional
(n5780) who underwent SLN mapping with lymphadenec- retrospective study concluded that SLN mapping versus
tomy versus lymphadenectomy alone, SLN mapping led to SLN mapping with lymphadenectomy in high-risk endo-
the detection of more metastasis (30.3% vs 14.7%, P,.001) metrial cancer did not impact survival outcomes.102 A re-
and was associated with greater use of adjuvant therapy.96 cent prospective, multicenter cohort study (SENTOR-
Long-term follow-up was reported from a prospective mul- trial) examined the diagnostic accuracy of SLN mapping
ticenter study in 125 patients with early-stage endometrial versus lymphadenectomy for intermediate- and high-
carcinoma who underwent SLN biopsy. Patients with a grade endometrial cancer in 156 patients. Of 27 patients

JNCCN.org | Volume 21 Issue 2 | February 2023 191


NCCN GUIDELINES® Uterine Neoplasms, Version 1.2023

with nodal metastasis, SLN mapping correctly identified SLN biopsy and ultrastaging detected metastatic SLNs
26 of them (96% sensitivity; 95% CI, 81%–100%), thus con- in a 3-fold greater number of patients than standard
cluding the acceptable accuracy of SLN mapping in high- lymphadenectomy.108
grade endometrial cancer.103 More studies have suggested The implications and appropriate management of
the value of using SLN mapping for surgical staging in micrometastases and isolated tumor cells (ITCs), jointly
high-grade endometrial cancer.104 referred to as low-volume metastases, detected via SLN
ultrastaging are not yet clear.109–112 Studies have recently
SLN Ultrastaging begun to investigate the significance of ITCs discovered
In general, SLN mapping allows for increased intraoperative during SLN mapping in early-stage endometrial cancer.
surgical precision to identify nodes more likely to harbor The AJCC 8th edition cancer staging manual indicates
metastasis combined with enhanced pathology protocols, that the lymph nodes with ITCs should be clearly reported
which has been shown to increase the detection of nodal even though they do not affect the overall staging.113
metastasis, which may alter stage and adjuvant therapy rec- When ITCs are detected in the absence of macrometasta-
ommendations. Studies have suggested that SLN ultrastag- sis and micrometastasis, the lymph node stage is desig-
ing leads to upstaging in 5% to 15% of patients.78,89,91,94,95 nated as pN0(i1).
Ultrastaging typically includes 2 components: serial A retrospective review examined 844 patients with
sectioning with review of multiple H&E-stained slides with endometrial cancer that underwent SLN mapping.114 The
or without cytokeratin IHC staining.105 Recent data highlight majority of patients with ITCs, micrometastasis, and mac-
the potential significance and impact of SLN ultrastaging to rometastasis received adjuvant chemotherapy (83%, 81%,
improve the accuracy of detecting micrometastases.106 and 89%, respectively). RFS at 3 years was 90% for those
In a cohort of 508 patients who underwent SLN map- with negative SLNs, 86% for ITCs, and 86% for micro-
ping, ultrastaging detected 23 additional cases of micro- metastasis. Only patients with SLN macrometastasis had
metastasis that would have been missed by conventional significantly lower RFS (71%, P,.001).
H&E staining.107 A multicenter study of 304 patients with A prospective observational study of 519 patients com-
presumed low- or intermediate-risk disease showed that pared outcomes for patients with SLN macrometastasis,

192 © JNCCN—Journal of the National Comprehensive Cancer Network | Volume 21 Issue 2 | February 2023
Uterine Neoplasms, Version 1.2023 NCCN GUIDELINES®

micrometastasis, and ITCs, taking into account adjuvant The LACE trial compared outcomes of patients with
treatment.115 Patients with SLN ITCs had a significantly bet- stage I endometrial carcinoma (n5760) who were random-
ter 3-year progression-free survival (PFS) compared with ized to undergo TH or total laparoscopic hysterectomy,
patients with SLN macrometastasis (95.5% vs 58.5%), and where half of the patients received concomitant lymphade-
outcomes were similar between patients with negative nectomy.118 At a median follow-up of 4.5 years, disease-free
SLNs, ITCs, and micrometastasis. Recurrence was detected survival (DFS) was 81.3% for laparotomy versus 81.6% for
in only 1 of 31 patients with ITCs (stage IB carcinosarcoma) laparoscopy, with no significant differences observed be-
and adjuvant treatment did not appear to influence out- tween groups for recurrence and OS. Another randomized
comes. Based on these early data, it is unclear if patients trial (n5283) comparing laparoscopy versus laparotomy re-
with SLN ITCs would derive significant benefit from adju- ported shorter hospital stay, less pain, and faster resumption
vant treatment.116 Future evaluation of prognosis/outcome of daily activities with laparoscopy.127 A recent follow-up
may need to prospectively examine the threshold for and study of a multicenter randomized trial evaluated outcomes
impact of adjuvant therapy for patients with scattered ITCs. for total laparoscopic hysterectomy versus TH in 279
patients with early-stage, low-risk endometrial cancer
Minimally Invasive Procedures who did not undergo concomitant lymphadenectomy
Over the past decade, practice has trended toward mini- and reported comparable disease recurrence and 5-year
mally invasive approaches to total abdominal hysterec- survival rates. The results were also similar to studies
tomy (TH)/BSO and lymph node assessment in patients with lymphadenectomy.128 Laparotomy may still be re-
with early-stage endometrial cancer.117 Although these quired for certain clinical situations (eg, elderly patients,
procedures may be performed by any surgical route (eg, those with a very large uterus) or certain metastatic
laparoscopic, robotic, vaginal, abdominal), the standard presentations.122,129,130
in those with apparent uterine-confined disease is to per- Robotic surgery is a minimally invasive technology
form the procedure via a minimally invasive approach. that has been increasingly used in the surgical staging
Randomized trials, a Cochrane Database Systematic Re- of endometrial carcinoma due to its potential advan-
view, and population-based surgical studies support that tages over laparotomy, especially for patients who are
minimally invasive techniques are preferred in the appro- overweight.131–135 Prospective cohort and retrospective
priate candidate due to a lower rate of surgical site infec- studies suggest that robotic approaches perform similarly
tion, transfusion, venous thromboembolism, decreased to laparoscopy and result in comparable or improved
hospital stay, and lower cost of care, without compromise perioperative outcomes.135–140 Oncologic outcomes
in oncologic outcome.117–123 Despite data showing that appear to be comparable to other surgical approaches,
minimally invasive procedures result in lower periopera- although longer-term outcomes (including quality of life)
tive complications and lower cost of care, racial and are still being investigated.141–143 In heavier patients,
geographic disparities in access to minimally invasive sur- robotic surgery may result in less frequent conversion
gical care have been observed.119,123 to laparotomy when compared with laparoscopic ap-
A randomized phase III trial evaluated laparoscopy for proaches and also appears to be safe and feasible in
comprehensive surgical staging; patients (n52,616) with patients at higher anesthesiologic risk.135,136,144
clinical stage I to IIA disease (GOG-LAP2) were assessed.124 Costs for robotic equipment and maintenance remain
Patients were randomly allocated 2:1 to laparoscopy or high.131,132,141–143,145,146 The SGO, American Association of
laparotomy. Results from LAP2 indicate that 26% of pa- Gynecologic Laparoscopists, and American Congress of
tients needed conversion to laparotomy because of poor Obstetricians and Gynecologists have published guidelines
visibility, metastatic cancer, bleeding, increased age, or in- or position statements about robotic surgery.147–149 For
creased body mass index. Detection of advanced cancer reviews on robotic-assisted surgery for gynecologic malig-
was not significantly different between the groups. How- nancies and associated cost issues, see Sinno and Fader150
ever, significant differences were noted in removal of pelvic and Gala et al.151
and para-aortic nodes (8% not removed with laparoscopy
vs 4% with laparotomy, P,.0001).125,126 Significantly fewer Primary Treatment
postoperative adverse events and shorter hospitalization These NCCN Guidelines divide pure endometrioid cancer
occurred with laparoscopy compared with laparotomy. Re- into 3 categories for delineating treatment: (1) disease lim-
currence rates were 11.4% for laparoscopy versus 10.2% for ited to the uterus; (2) suspected or gross cervical involve-
laparotomy. The 5-year overall survival (OS) rate was 84.8% ment; and (3) suspected extrauterine disease. Most patients
for both arms of LAP2.124 Laparoscopic staging was associ- with endometrial cancer have stage I disease at presenta-
ated with improved postoperative quality of life across sev- tion, and surgery (with or without adjuvant therapy) is rec-
eral parameters.121 ommended for medically operable patients. As a general

JNCCN.org | Volume 21 Issue 2 | February 2023 193


NCCN GUIDELINES® Uterine Neoplasms, Version 1.2023

principle, endometrial carcinoma should be removed en Fertility-Sparing Therapy


bloc to optimize outcomes; intraperitoneal morcellation Although the primary treatment of endometrial cancer is
should be avoided.152–155 usually hysterectomy, continuous progestin-based ther-
apy may be considered for highly selected patients with
Disease Limited to the Uterus grade 1, stage IA (noninvasive) disease who wish to pre-
To stage medically operable patients with endometrioid serve fertility.161–165 Likewise, it may also be selectively
histologies clinically confined to the fundal portion of used for young patients with endometrial hyperplasia
the uterus, the recommended surgical procedure in- who desire fertility preservation. The guidelines include
cludes removal of the uterus and bilateral tubes and ova- an algorithm for fertility-sparing therapy in selected
ries with lymph node and abdominal assessment (see patients with biopsy-proven grade 1 (preferably by
“Principles of Evaluation and Surgical Staging,” pages D&C), stage IA noninvasive endometrioid adenocarci-
186 and 187 and “Lymphadenectomy,” page 188, and noma (see “Criteria for Considering Fertility-Sparing
“Sentinel Lymph Node Mapping,” page 189).65 Ovarian Options,” available at NCCN.org). The panel recom-
preservation may be safe in select premenopausal pa- mends consultation with a fertility expert and genetic
evaluation of tumor and evaluation for inherited can-
tients with stage I endometrioid cancer.156–158 Minimally
cer risk. When considering fertility-sparing therapy, all
invasive surgery is the preferred approach when techni-
of the criteria must be met as outlined in the algorithm
cally feasible and is considered a quality measure by the
(eg, no metastatic disease) and a negative pregnancy
SGO and the American College of Surgeons (www.sgo.org/
test must be ensured. Patients should also receive
quality-outcomes-and-research/quality-indicators; https://
counseling that fertility-sparing therapy is not the
www.facs.org/quality-programs/cancer-programs/national-
standard of care for the treatment of endometrial car-
cancer-database/quality-of-care-measures). cinoma. TH/BSO with surgical staging is recom-
During surgery, the intraperitoneal structures should mended after childbearing is complete, if therapy is
be carefully evaluated, and suspicious areas should be not effective, or if progression occurs. Fertility-sparing
biopsied. While not specifically affecting staging, FIGO therapy is not recommended for high-risk patients (eg,
and AJCC recommend that peritoneal cytology should those with high-grade endometrioid adenocarcinomas,
be collected and results should be recorded. Cytology uterine serous carcinoma, clear cell carcinoma, carcino-
results should not be taken in isolation to guide adju- sarcoma, and leimyosarcoma).
vant therapy. Enlarged or suspicious lymph nodes Continuous progestin-based therapy may include
should be excised to confirm or rule out metastatic dis- megestrol acetate, medroxyprogesterone, or an intra-
ease. Retroperitoneal node dissection with pathologic uterine device containing levonorgestrel.161,162,166 A com-
evaluation—in the absence of clinically apparent plete response occurs in about 50% of patients.161 The
lymphadenopathy—is useful when using the 2009 use of progestin-based therapy should be carefully
FIGO staging criteria, but its routine use has been ques- considered in the context of other patient-specific
tioned (see “Lymphadenectomy,” page 188). For pa- factors, including contraindications such as breast
tients with stage II disease, TH/BSO is the standard cancer, stroke, myocardial infarction, pulmonary em-
procedure. Radical hysterectomy should only be per- bolism, deep vein thrombosis, and smoking. The
formed if needed to obtain negative margins. panel also recommends counseling for weight man-
Patients with apparent uterine-confined endome- agement and lifestyle modification (see “Healthy Life-
trial carcinoma are candidates for sentinel node map- styles” and “Nutrition and Weight Management” in
ping, which assesses the pelvic nodes bilaterally and the NCCN Guidelines for Survivorship, available at
may be less morbid than complete lymphadenectomy NCCN.org).
(see “Sentinel Lymph Node Mapping,” page 189). In patients receiving progestin-based therapies, the
Adherence to the NCCN SLN algorithm is critical. NCCN panel recommends close monitoring with endo-
metrial sampling (biopsies or D&C) every 3 to 6 months.
Incomplete Surgical Staging TH/BSO with staging is recommended (1) after childbear-
For patients with incomplete surgical staging and high- ing is complete; (2) if patients have documented progres-
risk intrauterine features, imaging is recommended, espe- sion on biopsy; or (3) if endometrial cancer is still present
cially in patients with higher grade histologies.159,160 Surgi- after 6 to 12 months of progestin-based therapy.165,167
cal restaging, including lymph node dissection, can also Although some young patients who had subsequent nega-
be done.69 Based on the imaging and/or surgical restaging tive endometrial biopsies after hormonal therapy were
results, recommended adjuvant treatment options are able to become pregnant (35%), the ultimate recurrence
provided in the algorithm (see “Adjuvant Treatment” of rate was high (35%).161,164,168,169 In patients with persistent
“Incompletely Surgically Staged,” available at NCCN.org). endometrial carcinoma after 6 months of failed hormonal

194 © JNCCN—Journal of the National Comprehensive Cancer Network | Volume 21 Issue 2 | February 2023
Uterine Neoplasms, Version 1.2023 NCCN GUIDELINES®

therapy, the panel recommends pelvic MRI to exclude levonorgestrel intrauterine device can also be considered
myoinvasion and nodal/ovarian metastasis before con- for select patients (eg, estrogen and progesterone receptor–
tinuing fertility-sparing therapy. positive [ER/PR-positive]). Patients receiving hormonal
In a study of premenopausal patients with stage IA therapy alone should be closely monitored by endome-
to B endometrial cancer, median 16-year follow-up data trial biopsy (eg, consider endometrial biopsies every
suggest that ovarian preservation is safe and not associ- 3–6 months).32,178
ated with an increased risk of cancer-related mortality.156 For suspected gross cervical involvement in disease
Other studies also suggest that ovarian preservation may not suited for primary surgery, EBRT and brachytherapy is
be safe in select patients.157 an effective treatment that can provide pelvic control and
long-term PFS (see “Principles of Radiation Therapy,” in
Suspected or Gross Cervical Involvement the NCCN Guidelines for Uterine Neoplasms, available at
For patients with suspected or gross cervical involvement NCCN.org).179–182 EBRT and brachytherapy may be admin-
(endometrioid histologies), cervical biopsy or pelvic MRI istered with or without platinum-based chemosensitiza-
should be performed if not done previously (see “Additional tion, depending on the clinical situation and medical
Workup,” page 183).159,160,170,171 If negative, patients are as- fitness of the patient. If rendered operable, local treatment
sumed to have disease that is limited to the uterus and are consisting of surgery should follow. Systemic therapy alone
treated as previously described, although a radical hysterec- is also a primary treatment option (category 2B), but
tomy may be performed when necessary to obtain negative
should be followed by EBRT plus brachytherapy if the pa-
margins. Distinguishing primary cervical carcinoma from
tient remains inoperable.
stage II endometrial carcinoma may be difficult. Thus, for
Patients with unresectable extrauterine pelvic dis-
patients suitable for primary surgery, TH or radical hyster-
ease (ie, vaginal, bladder, bowel/rectal, nodal, or parame-
ectomy is recommended along with BSO, cytology (perito-
trial involvement) are typically treated with EBRT with
neal lavage), and evaluation of lymph nodes if indicated
(or without) brachytherapy with (or without) systemic
(see “Principles of Evaluation and Surgical Staging,” page
therapy, followed by re-evaluation of tailored surgery.183–186
186).65 In these patients, radical or modified radical hys-
Systemic therapy alone can also be considered. Based on
terectomy may improve local control and survival when
treatment response, patients should be re-evaluated for
compared with TH.172,173 Alternatively, the patient may
surgical resection and/or RT.
undergo EBRT and brachytherapy (category 2B) followed
by TH/BSO and surgical staging.
Adjuvant Therapy
Suspected Extrauterine Disease Uterine-Confined Disease
If extrauterine disease (endometrioid histologies) is suspected, Thorough surgical staging provides important information
imaging studies are recommended along with CA-125 testing to assist in selection of adjuvant therapy for endometrial tu-
(see “Additional Workup” in the algorithm [page 184]). mors (see “Principles of Evaluation and Surgical Staging,”
ER testing is recommended in the setting of stage III or page 186). Patients with stage I endometrial cancer who
IV endometrioid tumors. Patients with no evidence of have thorough surgical staging are stratified by adverse risk
extrauterine disease are treated using the guidelines for factors (ie, age, positive LVSI, tumor size, and lower uterine
disease limited to the uterus. Patients with abdominal- segment or surface glandular involvement).187,188 Recom-
or pelvic-confined disease require surgical intervention mended adjuvant treatment is outlined in the algorithm
using TH/BSO with surgical staging and surgical debulk- (see the NCCN Guidelines for Endometrial Carcinoma at
ing with the goal to have no measurable residual disease; NCCN.org). Note that the treatment algorithm was revised
several studies support debulking.65,174,175 Consider preop- in 2010 based on the updated FIGO staging.46 However, by
erative chemotherapy.176 For distant visceral metastasis necessity, much of the discussion in this manuscript has
(eg, liver involvement), recommended options include been based on data from patients staged using the older
systemic therapy with (or without) EBRT with (or without) FIGO/AJCC staging system. The implications of stage migra-
TH/BSO and with (or without) SBRT. Ablative radiation tion should be considered when evaluating historical data.
can be considered for 1 to 5 metastatic lesions if disease is The basic concept underlying the recommendations
otherwise controlled (category 2B).177 in the NCCN Guidelines is the trend toward selection of
more aggressive adjuvant therapy for patients as tumor
Disease Not Suited for Primary Surgery grade and myometrial and/or cervical invasion increase,
For uterine-confined disease not suitable for primary sur- as risk of systemic metastases increase.189–191 In surgical
gery, EBRT and/or brachytherapy is the preferred treatment stage I and II endometrial cancer, other pathologic factors
approach. Alternatively, progestational agents (such as me- that may influence the decision regarding adjuvant ther-
droxyprogesterone acetate and megestrol acetate) and apy include LVSI, patient age, tumor volume, depth of

JNCCN.org | Volume 21 Issue 2 | February 2023 195


NCCN GUIDELINES® Uterine Neoplasms, Version 1.2023

invasion, and lower uterine segment or cervical glandular use of adjuvant RT improves pelvic control in patients
involvement. When administering adjuvant RT, it should with selected risk factors (and may improve PFS), but has
be initiated as soon as the vaginal cuff has healed, but no not been shown to improve OS. However, many of the
later than 12 weeks after surgery. earlier trials had limitations as the patients were primar-
Significant controversy centers on how much adju- ily low risk (ie, they had low-risk intrauterine pathologic
vant therapy is necessary in patients with surgical risk factors). It is recognized that in patients with uterine-
stage I endometrial cancer. The practice of surgical confined disease, there is a spectrum of risk based on
staging has led to a decrease in the use of adjuvant intrauterine pathologic findings. Adverse intrauterine
therapy for stage I endometrial carcinoma, which is re- pathologic risk factors include high-grade tumors, deep
flected in the option of observation in the NCCN myometrial invasion (and consequently more advanced
Guidelines for selected patients with low-risk features stage), LVSI (especially extensive), and serous or clear cell
(see section on “Adjuvant Treatment,” in the NCCN carcinoma histologies.
Guidelines for Endometrial Carcinoma, available at Four trials have evaluated the role of adjuvant
NCCN.org).74,188,189,192–194 The NCCN panel prefers obser- external-beam pelvic RT in patients with endometrial
vation for patients with stage IA, grade 1/2 disease, but carcinoma. In 2 of these trials, the patients were not
strongly suggests treatment with adjuvant vaginal brachy- formally staged (PORTEC-1,197 Aalders et al198). In the
therapy for those $60 years and/or those with LVSI. For third trial (ASTEC/EN.5), only 50% of the patients were
patients with stage IA, grade 3 tumors, especially in those thoroughly staged as part of a companion surgical pro-
who have been surgically staged, vaginal brachytherapy is tocol.63 However, formal surgical staging was mandated
the preferred option, or observation can be considered if for all patients in the GOG 99 trial.199 Note that these trials
no myometrial invasion is present. If higher risk factors used the older staging system (ie, before 2009).
are present, ie, age $70 years or LVSI, EBRT can be consid- The PORTEC-1 and GOG 99 trials suggest that external-
ered as a category 2B option. For patients with stage IB, beam pelvic RT provides a locoregional control benefit in
grade 1–2 disease, vaginal brachytherapy is preferred, al- selected patients with uterine-confined disease.197,200 Radia-
though observation can be considered if no adverse risk tion was not shown to increase OS.201 It is important to note
factors are present. In these patients, the Postoperative Ra- that the PORTEC 1 trial was powered to evaluate OS, al-
diation Therapy in Endometrial Carcinoma (PORTEC)-2 though the GOG 99 trial was not. Similarly the Aalders et al
trial, without evaluation of pelvic nodes, found pelvic re- randomized trial found that RT reduced vaginal (ie, locore-
currence to be low with vaginal brachytherapy alone.195 gional) recurrences but did not reduce distant metastases
EBRT can be considered in grade 2 tumors if additional or improve survival.198 A pooled randomized trial (ASTEC/
risk factors are present such as age $60 years and/or if EN.5) suggested that adjuvant pelvic RT alone did not im-
LVSI is present. For stage IB, grade 3 disease with adverse prove either relapse-free survival (ie, PFS) or OS in patients
risk factors, systemic therapy is added as a category 2B with intermediate-risk or high-risk early-stage endometrial
option (in addition to EBRT and/or vaginal brachytherapy). cancer, but there was an improvement in pelvic control.202
The recommended postoperative (ie, adjuvant) treat- However, the ASTEC/EN.5 study is very controversial: 51%
ment options for surgical stage II patients (using thorough of the patients in the ASTEC observation group received
surgical staging) are shown in the algorithm (see “Adjuvant vaginal brachytherapy. Vaginal brachytherapy has been
Treatment,” for stage II in the NCCN Guidelines for Endo- shown to decrease vaginal recurrence, and in PORTEC 2
metrial Carcinoma, at NCCN.org). The NCCN panel gener- vaginal brachytherapy was compared in a prospective ran-
ally agrees on the role of adjuvant therapy for patients with domized trial with EBRT in low-risk patients. Vaginal
an invasive cervical component if extrafascial hysterectomy brachytherapy alone was shown to sufficiently control the
is performed. However, for patients with stage II disease pelvis and was less toxic than full pelvic RT. As most pelvic
who have had a radical hysterectomy with negative surgical recurrences are vaginal, inclusion of vaginal brachytherapy
margins and no evidence of extrauterine disease, EBRT in the “observation” arm of the ASTEC/EN.5 study weak-
(preferred) and/or vaginal brachytherapy with (or without) ens any conclusions regarding pelvic radiation.67,203 The
systemic therapy (category 2B) are options. As with stage I GOG 99 trial showed that adjuvant pelvic RT improved lo-
disease, the presence of adverse risk factors (including coregional control and relapse-free interval (ie, PFS), with-
depth of stromal invasion, grade, LVSI, and adverse fundal out an OS benefit, although the study was not powered to
risk factors) should be considered when selecting adjuvant evaluate OS.199,204,205 In both trials, pelvic radiation was
therapy for stage II disease.196 found to be of greater benefit in patients .60 years with
higher grade and more deeply invasive disease.
Adjuvant RT To help select the appropriate patient population that
Several phase III trials have assessed adjuvant therapy in may benefit from adjuvant pelvic RT, the GOG 99 and
patients with uterine-confined disease. In summary, the PORTEC trials defined risk factors for patients at high-

196 © JNCCN—Journal of the National Comprehensive Cancer Network | Volume 21 Issue 2 | February 2023
Uterine Neoplasms, Version 1.2023 NCCN GUIDELINES®

intermediate risk (HIR) for recurrence, although the defini- brachytherapy plus chemotherapy arm compared with the
tion differed between these trials.197,199 Risk factors for re- pelvic EBRT arm. No significant between-group differences
currence identified in both trials included higher age, deep in vaginal or distant recurrence rates were observed. How-
myometrial invasion (.50%), higher grade (grade 2 or 3, ever, there were more extravaginal pelvic failures in the
serous or clear cell), and LVSI (especially extensive as de- brachytherapy plus chemotherapy arm. At a median fol-
fined in the PORTEC trials). Based on risk factors identified low-up of 53 months, 3-year RFS was 82% for both treat-
in GOG 99, HIR disease was defined as disease in patients ment arms; 3-year OS was 88% for the brachytherapy plus
,50 years with grade 2 or 3 disease, myometrial invasion chemotherapy cohort and 91% for the pelvic EBRT cohort.
greater than 50%, and LVSI.199 Patients 50 to 70 years of Acute toxicity was more common and severe for patients
age were considered HIR if they had 2 of the 3 identified receiving brachytherapy with chemotherapy. No differences
high-risk features. Patients $70 years were defined as HIR in late-onset toxicities were observed.214 Questions were
if they also had one risk feature present. Based on data raised whether 3 cycles of chemotherapy were sufficient to
from PORTEC-1, HIR patients were defined as having 2 of control distant disease.
3 risk factors (ie, age .60 years, deep myometrial invasion, Analysis of pooled data from PORTEC-1 and PORTEC-2
grade 3 histology).197,204 LVSI was not considered in the ranked the predictive power of multiple variables on
original PORTEC trials, but a subsequent retrospective patient outcomes examined in these trials. Patient age,
evaluation demonstrated increased recurrence with exten- tumor grade, and LVSI were highly predictive for locore-
sive LVSI, as defined by the protocol. gional relapse, distant relapse, OS, and DFS, and treatment
Due to concerns about potential toxicity of external- given (EBRT vs vaginal brachytherapy) was predictive for
beam pelvic RT, the role of vaginal brachytherapy alone locoregional relapse and DFS.187 The benefit of adjuvant
in uterine-confined disease has been evaluated. PORTEC-2 EBRT in the highest risk spectrum of uterine-confined dis-
randomly assigned patients to external-beam pelvic RT ease remains controversial. Most NCCN panel members
versus vaginal brachytherapy alone in uterine-confined feel that patients with deeply invasive grade 3 tumors
disease. PORTEC-2 showed excellent and equivalent vagi- should receive adjuvant treatment. Two large retrospective
nal and pelvic control rates with both adjuvant radiation SEER analyses of patients with endometrial cancer found
approaches and no difference in OS.195 Given that vaginal that adjuvant RT improved OS in those with high-risk dis-
brachytherapy is associated with significantly less toxicity ease.215,216 In a meta-analysis of randomized trials, a subset
than pelvic RT, vaginal brachytherapy alone is a reason- analysis found that adjuvant pelvic RT for stage I disease
able choice for patients with uterine-confined endome- was associated with a trend toward a survival advantage in
trial cancer as defined in the PORTEC 2 trial.195,204–212 the highest-risk spectrum (eg, those with 1988 FIGO stage
The use of vaginal brachytherapy and/or whole pelvic RT IC grade 3) but not in lower risk patients; however, other
should be carefully tailored to a patient’s pathologic find- reviews have shown conflicting results.207,217–221
ings. Both PORTEC-1 and PORTEC-2 specifically excluded The long-term follow-up study (median 20.5 years) of
patients with 1998 FIGO stage 1C and grade 3 endometrial 568 patients with early-stage endometrial carcinoma en-
carcinoma (2009 FIGO stage IB, grade 3)46; thus, the use rolled in the Aalders trial compared long-term outcomes in
of adjuvant brachytherapy alone in this higher risk subset patients who received vaginal brachytherapy plus EBRT
remains more controversial. PORTEC studies did not eval- versus vaginal brachytherapy alone. The findings suggested
uate lymph nodes and, therefore, in the context of com- no statistical difference in OS between the study groups,
plete surgical staging and the lack of a survival benefit, and in this cohort, patients ,60 years of age who received
the need for pelvic irradiation remains controversial in EBRT had increased incidence of secondary cancers and
uterine-confined disease. subsequent higher mortality rates.207 Evaluation of second-
A meta-analysis evaluated results from studies that ary malignancies in the context of increased genetic sus-
compared adjuvant postoperative EBRT with or without ceptibility (eg, MSI-H) and radiation is ongoing.
vaginal brachytherapy and vaginal brachytherapy alone in
stage II endometrial cancer. EBRT plus vaginal brachy- Adjuvant Systemic Therapy
therapy significantly reduced locoregional recurrence ver- Patients with deeply invasive, grade 3, uterine-confined dis-
sus vaginal brachytherapy alone. OS was comparable in ease (2009 FIGO stage IB, grade 3 [formerly 1988 FIGO stage
both arms.213 IC, grade 3]) have a relatively poor prognosis. Despite adju-
The GOG 249 trial examined vaginal cuff brachyther- vant therapy with pelvic RT, a significant number of patients
apy and 3 cycles of carboplatin/paclitaxel therapy (3 cycles) continue to have a significant risk of distant metastases, and
versus pelvic EBRT only in patients with high-risk, uterine- an optimal adjuvant therapy is still sought.199,200 Therefore,
confined endometrial carcinoma (n5601), including serous some clinicians suggested that adding systemic therapy to
and clear cell carcinoma. GOG 249 reported significantly adjuvant RT may provide added therapeutic benefit (ie, de-
increased rates of nodal recurrence (primarily pelvic) in the crease in distant metastases).189,222 Studies have evaluated

JNCCN.org | Volume 21 Issue 2 | February 2023 197


NCCN GUIDELINES® Uterine Neoplasms, Version 1.2023

the role of systemic therapy in highest risk uterine-confined patients with extrauterine disease. Thus, in the NCCN
disease.222,223 PFS is improved with adjuvant sequential che- Guidelines, systemic therapy forms the established frame-
motherapy.222 However, the NCCN panel feels that adjuvant work of adjuvant therapy for patients with stage III or IV
systemic therapy is a category 2B recommendation in this disease. Whole abdominal RT as a single modality (as
setting because an OS advantage has not been shown.222 The used in GOG 122) is considered inferior to chemotherapy
GOG-249 phase 3 trial evaluated the benefit of adjuvant and is too toxic; therefore, it is no longer recommended.
pelvic RT versus vaginal cuff brachytherapy plus 3 cycles For the purposes of these guidelines, whole abdominal RT
of paclitaxel/carboplatin combination in 601 patients is not considered to be tumor-directed RT (see “Principles
with high-intermediate and high-risk early-stage endo- of Radiation Therapy,” in the NCCN Guidelines for Uter-
metrial cancer. The 5-year RFS and OS were similar in ine Neoplasms at NCCN.org).
both groups and superiority of any of these treatments Recurrences were frequent in both treatment arms
were not demonstrated. Acute toxicity was greater in the of GOG 122, occurring in the pelvis and abdomen. Ap-
combination therapy.214 proximately 52% of patients with advanced endometrial
carcinoma had recurrences, indicating the need for fur-
Advanced Stage/Extrauterine Disease ther therapeutic improvement in this high-risk popula-
A consensus exists that patients with documented extra- tion.184 A study found that combined modality adjuvant
uterine disease are at increased risk for recurrence and therapy (using chemotherapy and tumor-directed RT)
need adjuvant therapy; however, the optimal form of ad- may provide a therapeutic benefit when compared with
juvant therapy has yet to be determined.224,225 Patients single-modality adjuvant therapy.186,232,233
with extrauterine disease confined to the lymph nodes or A follow-up study evaluated the role of chemotherapy
the adnexa may be treated with pelvic or extended-field “intensification” for this patient population. The GOG
RT alone or with chemotherapy (radiation is targeted to 184 trial compared 2 chemotherapy regimens (cisplatin
sites of nodal disease).226 However, systemic therapy is and doxorubicin with or without paclitaxel) with tumor-
regarded as the foundation of adjuvant therapy for pa- directed radiation (involved-field radiation either to the
tients with extrauterine disease. The NCCN Guidelines pelvis or to the pelvis plus para-aortic nodes). Results in-
include carboplatin/paclitaxel as the preferred option in dicate that the 3-drug regimen did not improve survival
the primary/adjuvant setting for advanced-stage disease when compared with the 2-drug regimen after 3 years of
or high-risk histologies.227–229 follow-up and that the more intensive chemotherapy re-
For stages III and IV disease, systemic therapy sulted in greater toxicity (eg, hematologic toxicity, sensory
forms the mainstay of treatment and can be combined neuropathy, myalgia).185
with EBRT with (or without) vaginal brachytherapy. In a retrospective review of 116 patients with stage IIIC
The combination of therapies depends on assessment endometrial cancer, adjuvant RT significantly improved
of both locoregional and distant metastatic risk. Com- OS in patients with endometrioid histology, high-grade tu-
bination therapy can be considered for stages IIIB and mors, and positive para-aortic lymph nodes. Conversely,
IIIC disease. patients with low-grade tumors and nonendometrioid his-
Previously, whole abdominal RT was used for carefully tology who received RT had similar OS compared with
selected patients deemed at risk for peritoneal failure, and those who did not.234 In a multicenter retrospective review
RT appeared to have provided therapeutic benefit in retro- of 73 patients with stage IIIA endometrial carcinoma, sur-
spective studies. However, it is considered too toxic and gery followed by both chemotherapy and RT provided the
has largely been abandoned.230,231 A randomized phase III highest 5-year OS.235 A prospective study of 122 patients
GOG (122) trial assessed optimal adjuvant therapy for with fully resected locally advanced disease suggested a po-
patients with endometrial cancer who had extrauterine tential benefit of adjuvant chemoradiation followed by che-
disease. In this trial, patients with stage III and intra- motherapy, with an estimated 5-year PFS and OS of 73%
abdominal stage IV disease who had minimal residual dis- and 84%.236 Adjuvant therapy options were compared in a
ease were randomly assigned to whole abdominopelvic multicenter retrospective analysis of 265 patients with opti-
RT versus 7 cycles of combined doxorubicin (60 mg/m2) mally resected stage IIIC endometrial carcinoma. Com-
and cisplatin (50 mg/m2) treatment, with an additional cy- pared with patients receiving adjuvant RT or adjuvant RT
cle of cisplatin (AP). This GOG trial reported that AP che- plus chemotherapy, patients who received adjuvant che-
motherapy improved PFS and OS when compared with motherapy alone had a 2.2-fold increased risk of recurrence
whole abdominopelvic RT; however, acute toxicity (eg, and a 4.0-fold increased risk of death.225
peripheral neuropathy) was greater in the AP chemother- Multimodality therapy is now the basis of randomized
apy arm.184 trials evaluating therapy. The phase 2, RTOG 9708 trial as-
The GOG 122 study established the role of adjuvant sessed 46 patients for safety, toxicity, recurrence, and survival
multiagent systemic chemotherapy for curative intent in when chemotherapy (cisplatin/paclitaxel) was combined

198 © JNCCN—Journal of the National Comprehensive Cancer Network | Volume 21 Issue 2 | February 2023
Uterine Neoplasms, Version 1.2023 NCCN GUIDELINES®

with adjuvant radiation in patients with high-risk endome- patients with no prior RT exposure at the recurrence site,
trial cancer. The trial participants included patients with the panel recommends EBRT with (or without) brachy-
grade 2 or 3 endometrial adenocarcinoma with either .50% therapy and systemic therapy, or surgery with (or without)
myometrial invasion, cervical stromal invasion, or pelvic- intraoperative RT (IORT) and systemic therapy. For pa-
confined extrauterine disease. The OS and DFS favored the tients previously treated with brachytherapy only at the
combined modality treatment.237 recurrence site, surgery with (or without) IORT is recom-
The phase III, PORTEC-3 trial investigated the benefit mended (category 3 for IORT).
of combined adjuvant chemotherapy and EBRT versus For patients previously treated with EBRT at the re-
EBRT alone in 686 patients with endometrial cancer (stage currence site, recommended therapy for isolated relapse
I, grade 3 with deep invasion, LVSI, or both; stage II; stage includes surgery with (or without) IORT (category 3 for
III; or any patient with stage I to III serous or clear cell en- IORT) 6 systemic therapy. Use of RT in the context of re-
dometrial cancer). The 5-year OS was 81.4% (95% CI, currence depends on the site of recurrence (inside or
77.2–85.8) with chemoradiotherapy versus 76.1% (95% outside the prior radiation field), and dose of prior ther-
CI, 71.6–80.9) with radiotherapy alone (hazard ratio apy. Reirradiation is used only in the context of limited
[HR], 0.70; 95% CI, 0.51–0.97; P5.034) and 5-year failure- disease for palliation and lack of other options. In se-
free survival was 76.5% (95% CI, 71.5–80.7) versus 69.1% lected patients, radical surgery (ie, pelvic exenteration)
(63.8–73.8; HR, 0.70; 95% CI, 0.522–0.94; P5.016).238 Patients has been performed with reported 5-year survival rates
with serous cancers and with stage III disease were shown approximating 20%.243–246
to benefit the most from the addition of systemic therapy. Isolated vaginal recurrences treated with RT have good
The combination treatment was also shown to be associ- local control and 5-year survival rates of 50%–70%.247,248
ated with more severe adverse events.239 Prognosis is worse if there is extravaginal extension or pel-
The GOG-258 phase III trial evaluated 707 patients vic lymph node involvement.247 After RT, it is unusual for
with stage III or IVA, high-risk endometrial cancer who patients to have recurrences confined to the pelvis. The
were randomly assigned 1:1 to receive chemoradiother- management of such patients remains controversial.
apy or chemotherapy only.240 This trial supported the Additional therapy options for disease confined to
benefit of using chemotherapy alone by concluding that vagina or paravaginal soft tissues include EBRT with (or
the combined therapy was not associated with longer re- without) brachytherapy with (or without) systemic ther-
lapse-free survival when compared with chemotherapy apy. EBRT and systemic therapy are also included as op-
alone (59% vs 58%, respectively). OS results are pending. tions for the additional treatment of pelvic lymph node
A follow-up molecular analysis was performed on recurrence, para-aortic or common iliac lymph node in-
the PORTEC-3 trial to study the impact of chemoradio- vasion, and upper abdominal or peritoneal recurrences
therapy for each molecular subtype using tissue samples as shown in the algorithm (see “Additional Therapy,” in
from the trial participants. The tumors were classified the algorithm, page 185).
into p53 abnormal, POLE, MMR-deficient (dMMR), or no
specific molecular profile. The 5-year RFS with chemora- Distant Metastases
diotherapy versus RT alone was p53 abnormal, 59% ver- For gross upper abdominal residual disease, more aggres-
sus 36%; POLE, 100% versus 97%; dMMR, 68% versus sive treatment of relapse is recommended, as outlined for
76%; and 80% versus 68% for no specific molecular pro- disseminated metastases in “Therapy for Relapse” in the
file, suggesting that systemic therapy was beneficial for algorithm (page 185). For resectable isolated metastases,
consider surgical resection and/or EBRT, or ablative ther-
those patients with the p53 abnormal tumors.241 Results
apy. Ablative RT can be considered for 1 to 5 metastatic
are awaited for an ongoing PORTEC-4a trial investigating
lesions if the primary cancer has been controlled (cate-
molecular profile-based directed adjuvant treatment in
gory 2B).177 Providers can also consider systemic therapy
high-risk endometrial cancer.242
(category 2B). Further recurrences or disease not amena-
ble to local therapy are treated as disseminated metasta-
Treatment of Recurrent or Metastatic Disease
ses. Treatment options for disseminated metastases are
Locoregional Recurrence systemic therapy with (or without) palliative EBRT. For
Patients with local or regional recurrences (negative for persistent progression of disseminated metastases, best
distance metastases on radiologic imaging) can be evalu- supportive care is recommended (see the NCCN Guide-
ated for further treatment (see “Clinical Presentation,” in lines for Palliative Care at NCCN.org).
the algorithm [page 185]). For recurrences confined to the
vagina or the pelvis alone, second-line treatment (typi- Hormonal Therapy
cally with RT and/or surgery or systemic therapy) can be The role of hormonal therapy in recurrent or metastatic
effective, and selection depends on prior therapy. For cancer has been primarily evaluated in patients with

JNCCN.org | Volume 21 Issue 2 | February 2023 199


NCCN GUIDELINES® Uterine Neoplasms, Version 1.2023

endometrioid histologies only. Hormonal therapy is typically Systemic Therapy


used for lower-grade endometrioid histologies, preferably in Chemotherapy for endometrial cancer has been exten-
patients with small tumor volume or an indolent growth sively studied.264,265 Based on the current data, multiagent
pace. Hormonal agents for treating recurrent/metastatic dis- chemotherapy regimens are preferred for advanced dis-
ease include megestrol acetate with alternating tamoxifen, ease, if tolerated. The NCCN Guidelines for Endometrial
medroxyprogesterone acetate/tamoxifen (alternating), ever- Carcinoma recently updated the systemic therapy recom-
olimus/letrozole combination, progestational agents (such mendation by including multiagent chemotherapy regi-
as medroxyprogesterone acetate and megestrol acetate), aro- mens such as carboplatin/paclitaxel, carboplatin/docetaxel,
matase inhibitors, tamoxifen alone, or fulvestrant.249–254 No and carboplatin/paclitaxel/bevacizumab as first-line ther-
particular drug, dose, or schedule has been found to be supe- apy options for the recurrent disease setting.
rior. The main predictors of response in the treatment of Other combination therapies such as cisplatin/
metastatic disease are well-differentiated tumors, expression doxorubicin, cisplatin/doxorubicin/paclitaxel, ifosfamide/
of ER/PR receptors, a long disease-free interval, and the loca- paclitaxel (for carcinosarcoma), cisplatin/ifosfamide (for
tion and extent of extrapelvic (particularly pulmonary) carcinosarcoma) are added as second-line or subsequent-
metastases. therapy options. A phase III randomized trial (GOG 177)
For asymptomatic or low-grade disseminated metas- compared 2 combination chemotherapy regimens in
tases, hormonal therapy with progestational agents has females with advanced or recurrent endometrial carci-
shown good responses, particularly in patients with ER/ noma. The 273 participants were randomly assigned
PR-positive disease.255–258 Tamoxifen has a 20% response to (1) cisplatin/doxorubicin/paclitaxel; or (2) cisplatin/
rate in disease that does not respond to standard proges- doxorubicin. The 3-drug regimen was associated with
terone therapy.259 Tamoxifen has also been combined improved survival (15 vs 12 months; P,.04) but with sig-
with progestational agents; however, a few patients had nificantly increased toxicity (ie, peripheral neuropathy);
grade 4 thromboembolic events with this combination therefore, it is not widely used.266–268 These regimens are
regimen.251,252,260 In some patients, aromatase inhibitors recommended as second-line options in the NCCN Guide-
(eg, anastrozole, letrozole) may be substituted for proges- lines, because most panel members feel that carboplatin/
tational agents or tamoxifen.257 paclitaxel is a less toxic and preferred first-line option. The
Everolimus combined with letrozole is recommended response rates with other multiagent chemotherapies have
for recurrent disease of endometrioid histology. In the ranged from 31% to 81%, but with relatively short dura-
phase II trial, in patients with progressive or recurrent en- tions. The median survival for patients in such trials re-
dometrial cancer who had received up to 2 prior thera- mains approximately 1 year.264,265
pies, the clinical benefit rate and objective response rate Carboplatin and paclitaxel is an increasingly used
among 35 evaluable patients was 40% and 32%, respec- regimen for advanced or recurrent endometrial cancer;
tively.261 In a following phase II study, patients (with or the response rate is about 40% to 62%, and OS is about 13
without prior chemotherapy) were treated either with the to 29 months.269–271 A phase III trial (GOG 209) compared
everolimus/letrozole combination or medroxyprogester- carboplatin and paclitaxel versus cisplatin, doxorubicin,
one acetate/tamoxifen regimen. Twenty-two percent of paclitaxel, and filgrastim (granulocyte colony-stimulating
patients responded to the everolimus/letrozole therapy, factor). Trial data show that oncologic outcomes are similar,
while 25% showed a response with the medroxyproges- but the toxicity and tolerability profile favor carboplatin/
terone acetate/tamoxifen regimen.262 Median PFS was paclitaxel.272 Thus, the carboplatin/paclitaxel regimen is
6 months for the everolimus/letrozole arm and 4 months now the preferred approach for many patients. For patients
for the hormonal therapy arm. Median OS was 31 months in whom paclitaxel is contraindicated, docetaxel can be
and 17 months for the everolimus/letrozole and medroxy- considered in combination with carboplatin.273
progesterone acetate/tamoxifen arms, respectively. Higher A phase II trial initially examined the addition of
PFS was observed in both arms for patients who had not bevacizumab to carboplatin and paclitaxel among 15
received any prior chemotherapy. patients with advanced or recurrent endometrial carci-
Other hormonal modalities have not been well-stud- noma.274 Although this study was closed early due to
ied, and adjuvant therapy with hormonal agents has not the initiation of a national trial, a retrospective analysis
been compared with cytotoxic agents.257,263 If disease pro- was performed to include data from an additional 27
gression is observed after hormonal therapy, cytotoxic che- patients who had received carboplatin/paclitaxel/beva-
motherapy can be considered. However, clinical trials or cizumab for advanced or recurrent disease.275 Collec-
best supportive care (see the NCCN Guidelines for Palliative tive median PFS was 20 months with a median OS of 56
Care, available at NCCN.org) are appropriate for patients months. An overall response rate (ORR) of 82.8% was
with disseminated metastatic recurrence who have a poor noted, with an 87.5% response rate among the subset
response to hormonal therapy and chemotherapy. of 8 patients who received this triplet as second-line

200 © JNCCN—Journal of the National Comprehensive Cancer Network | Volume 21 Issue 2 | February 2023
Uterine Neoplasms, Version 1.2023 NCCN GUIDELINES®

therapy after carboplatin/paclitaxel.275 Another phase II TMB-H testing if not previously done and has included
randomized study showed that the carboplatin/pacli- pembrolizumab as a category 1 option for patients with un-
taxel/bevacizumab combination improved OS from resectable or metastatic tumors with TMB-H (.10 mut/Mb),
29.7 to 40 months compared with the doublet regi- as determined by a validated and/or FDA-approved test,
men.276 Another meta-analysis of 3 studies also con- who have progressed after prior treatment and who have no
cluded similar results where the triplet combination satisfactory alternative treatment options.
increased the OS and PFS at .12 months with an ORR Further studies have indicated that pembrolizumab
of 76%.277 monotherapy is less active in patients with MSI-stable or
If multiagent chemotherapy regimens are contraindi- MMR-proficient (pMMR) disease versus MSI-H/dMMR dis-
cated, then single-agent subsequent-line therapy options ease. Only 16% to 31% of endometrial cancers are MSI-H/
included in the guidelines are cisplatin, carboplatin, doxo- dMMR.286,292,293 The Keynote-146 phase I/II trial showed
rubicin, liposomal doxorubicin, paclitaxel, albumin-bound that the combination of pembrolizumab/lenvatinib had a
paclitaxel, topotecan, cabozantinib, and docetaxel (category promising antitumor response in patients with advanced
2B for docetaxel).257,258,278–281 When single agents are used endometrial cancer regardless of their tumor MSI status.294
as second-line treatment, responses range from 4% to 27%; The Keynote-775 phase III trial randomly assigned 827 pa-
paclitaxel is the most active in this setting.281 Some oncolo- tients with pMMR (MSI-stable) advanced endometrial can-
gists have used liposomal doxorubicin, because it is less cer to receive pembrolizumab/lenvatinib combination or
toxic than doxorubicin; the response rate of liposomal chemotherapy (doxorubicin or paclitaxel).295 The median
doxorubicin is 9.5%.282 Docetaxel is recommended for use PFS for the pembrolizumab/lenvatinib arm was 7.2 versus
as a single agent; however, it is a category 2B recommenda- 3.8 months for the chemotherapy arm (HR, 0.56; 95% CI,
tion because it is less active (7.7% response rate) than other 0.47–0.66; P,.001). The median OS was also longer for the
agents.283,284 Bevacizumab was shown to have a 13.5% re- pembrolizumab/lenvatinib arm than for the chemotherapy
sponse rate and OS rate of 10.5 months in a phase II trial arm (18.3 vs 11.4 months; HR, 0.62; 95% CI, 0.51–0.75;
for persistent or recurrent endometrial cancer.285 Based on P,.001). Based on these data, the NCCN Guidelines in
these studies, the NCCN panel considers bevacizumab as
Endometrial Carcinoma include lenvatinib/pembrolizumab
appropriate single-agent biologic therapy for patients who
as a category 1 option for pMMR tumors for patients who
have progressed on previous cytotoxic chemotherapy.285
have received prior platinum-based therapy.
In the advanced endometrial cancer cohort (n524) of
Other anti-PD-1 inhibitors, such as dostarlimab and
the phase Ib KEYNOTE-028 trial, durable antitumor re-
nivolumab, have also shown antitumor activity against
sponses were noted in a small subset of patients with pro-
MSI-H tumors. Dostarlimab is being evaluated in the
grammed death ligand 1 (PD-L1)–positive tumors (3 partial
ongoing GARNET phase I trial for patients with advanced
response, 3 stable disease).286 Studies have also indicated
endometrial cancer with dMMR/MSI-H disease. The ORR
that dMMR tumors are sensitive to programmed death
after 16.3 months was 43.5% with manageable safety pro-
receptor-1 (PD-1) blockade.287–289 Results were published
from a study of patients with dMMR tumors of various dis- file. Dostarlimab is approved as monotherapy for the treat-
ease sites. Among patients with dMMR endometrial carci- ment of patients with recurrent or advanced dMMR/MSI-H
noma who received pembrolizumab (n515), the objective endometrial cancer that has progressed on or after prior
response rate was 52% and the disease control rate was treatment with a platinum-containing regimen. Nivolumab
73% (3 complete response, 5 partial response, and 3 stable monotherapy has also shown promising activity in endo-
disease).287 The FDA expanded pembrolizumab approval in metrial carcinoma with dMMR tumors.296 The PD-L1 inhib-
2017 to include treatment of unresectable or metastatic, itor, avelumab, have shown an ORR of 26.7% in advanced
MSI-H, or dMMR solid tumors that have progressed after endometrial cancer with dMMR tumor as monotherapy.
prior treatment and that have no satisfactory alternative Dostarlimab, nivolumab, and avelumab are included as
treatment options.289 In the following phase II Keynote-158 biomarker-directed options for recurrent dMMR/MSI-H
trial, pembrolizumab further demonstrated robust antitu- endometrial tumors for those patients who have received
mor activity with encouraging survival outcomes in patients prior systemic therapy. The NCCN panel also recommends
with advanced MSI-H/dMMR endometrial cancer and larotrectinib or entrectinib for NTRK gene fusion-positive,
manageable adverse events.290 Pembrolizumab is included recurrent/advanced endometrial tumors as a category 2B
as a category 1 treatment option for MSI-H/dMMR endo- option.
metrial tumors with a recommendation that recurrent en- The systemic therapy options for high-risk histolo-
dometrial tumors be tested for MSI-H or dMMR if not done gies recommended in the NCCN Guidelines include car-
previously. The FDA has also authorized the use of pembro- boplatin/paclitaxel as a preferred, category 1 option for
lizumab in TMB-high tumors (.10 mutations/megabase patients with carcinosarcoma histology. A randomized
[mut/Mb]).291 For TMB-H tumors, the panel recommends phase II study examined the addition of trastuzumab to

JNCCN.org | Volume 21 Issue 2 | February 2023 201


NCCN GUIDELINES® Uterine Neoplasms, Version 1.2023

carboplatin/paclitaxel for patients with advanced or re- ifosfamide and paclitaxel increased survival and was less
current HER2/neu-positive uterine serous carcinoma.297 toxic than the previously used cisplatin/ifosfamide regi-
Among patients with stage III/IV disease undergoing men.299,301 OS was 13.5 months with ifosfamide/paclitaxel
primary treatment (n541), median PFS was 17.9 versus versus 8.4 months with ifosfamide alone.299,302
9.3 months for the experimental and control arms, re-
spectively (P5.013). PFS for patients with recurrent dis- Summary
ease (n517) was 9.2 versus 6.0 months (P5.003). The The management of endometrial cancer involves a
addition of trastuzumab appeared to improve PFS with- multimodality treatment comprised of surgery and/or
out increasing overall toxicity. The safety and tolerability systemic therapy and radiotherapy depending on the
of the trastuzumab combination was further evaluated in histopathologic assessment of the tumor, and clinical
61 patients in a recent phase II trial with PFS as the presentation. The prognosis of the disease is mainly
primary endpoint.298 The triplet therapy regimen carbo- assessed by histopathologic characteristics including
platin/paclitaxel/trastuzumab is recommended by the the grade, type, LVSI, and myometrial invasion. The
NCCN panel as a preferred option for HER2-positive NCCN Guidelines for Endometrial Cancer also include
uterine serous carcinoma or HER2-positive carcinosar- recommendations for molecular characterization of
coma as: (1) primary therapy for stage III/IV disease; or the tumor and encourages comprehensive genomic
(2) a first-line option for recurrent disease. The NCCN profiling, if feasible. The surgical staging of the disease
panel has designated the regimen a category 2B option impacts the prognosis and guides adjuvant treatment
for HER2-positive carcinosarcoma in both disease set- decisions, and is an integral part of management of
tings. This triplet regimen is recommended for patients the disease. Although surgery and/or radiotherapy is
who have not received any prior trastuzumab therapy. In typically the standard procedure to treat early-stage
second-line/subsequent therapy, the NCCN panel has in- endometrial cancer, more advanced stages or high-
cluded ifosfamide, ifosfamide/paclitaxel, and ifosfamide/ risk histologies of endometrial cancer are managed by
cisplatin as options for carcinosarcoma only. For treating a combination of radiotherapy and systemic therapy.
carcinosarcoma, ifosfamide was historically considered The appropriate therapy also takes into account the
the most active single agent.299,300 A phase III trial for ad- clinical situation with consideration of the patient’s
vanced carcinosarcoma showed that the combination of goals of care, especially in more advanced stages.

References
1. Siegel RL, Miller KD, Fuchs HE, et al. Cancer statistics, 2022. CA Cancer 13. Raffone A, Travaglino A, Raimondo D, et al. Prognostic value of myo-
J Clin 2022;72:7–33. metrial invasion and TCGA groups of endometrial carcinoma. Gynecol
2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin Oncol 2021;162:401–406.
2016;66:7–30. 14. Droegemueller W. Diagnostic procedures. Imaging, endometrial sam-
3. Van den Bosch T, Coosemans A, Morina M, et al. Screening for uterine pling, endoscopy: indications and contraindications, complications. In:
tumours. Best Pract Res Clin Obstet Gynaecol 2012;26:257–266. Katz VL, Lentz GM, Lobo RA, et al, eds. Comprehensive Gynecology.
5th ed. Mosby; 2007.
4. Kitchener HC, Trimble EL. Endometrial cancer state of the science
meeting. Int J Gynecol Cancer 2009;19:134–140. 15. Resnick KE, Hampel H, Fishel R, et al. Current and emerging trends in
Lynch syndrome identification in women with endometrial cancer.
5. Dinkelspiel HE, Wright JD, Lewin SN, et al. Contemporary clinical man- Gynecol Oncol 2009;114:128–134.
agement of endometrial cancer. Obstet Gynecol Int 2013;2013:583891.
16. McAlpine JN, Chiu DS, Nout RA, et al. Evaluation of treatment effects
6. Obermair A, Youlden DR, Young JP, et al. Risk of endometrial cancer in patients with endometrial cancer and POLE mutations: an individual
for women diagnosed with HNPCC-related colorectal carcinoma. Int J patient data meta-analysis. Cancer 2021;127:2409–2422.
Cancer 2010;127:2678–2684.
17. Jumaah AS, Salim MM, Al-Haddad HS, et al. The frequency of POLE-
7. Siegel RL, Miller KD, Jemal A. Cancer Statistics, 2017. CA Cancer J Clin mutation in endometrial carcinoma and prognostic implications: a sys-
2017;67:7–30. temic review and meta-analysis. J Pathol Transl Med 2020;54:471–479.
8. Ueda SM, Kapp DS, Cheung MK, et al. Trends in demographic and 18. Travaglino A, Raffone A, Stradella C, et al. Impact of endometrial carci-
clinical characteristics in women diagnosed with corpus cancer and their noma histotype on the prognostic value of the TCGA molecular sub-
potential impact on the increasing number of deaths. Am J Obstet groups. Arch Gynecol Obstet 2020;301:1355–1363.
Gynecol 2008;198:218.E1–E6. 19. Kwon JS, Scott JL, Gilks CB, et al. Testing women with endometrial
9. Chan JK, Sherman AE, Kapp DS, et al. Influence of gynecologic oncolo- cancer to detect Lynch syndrome. J Clin Oncol 2011;29:2247–2252.
gists on the survival of patients with endometrial cancer. J Clin Oncol 20. Buchanan DD, Tan YY, Walsh MD, et al. Tumor mismatch repair
2011;29:832–838. immunohistochemistry and DNA MLH1 methylation testing of patients
10. Benedetti Panici P, Basile S, Salerno MG, et al. Secondary analyses from with endometrial cancer diagnosed at age younger than 60 years opti-
a randomized clinical trial: age as the key prognostic factor in endome- mizes triage for population-level germline mismatch repair gene muta-
trial carcinoma. Am J Obstet Gynecol 2014;210:363.E1–E10. tion testing. J Clin Oncol 2014;32:90–100.
11. Doll KM, Tseng J, Denslow SA, et al. High-grade endometrial cancer: 21. Ferguson SE, Aronson M, Pollett A, et al. Performance characteristics of
revisiting the impact of tumor size and location on outcomes. Gynecol screening strategies for Lynch syndrome in unselected women with
Oncol 2014;132:44–49. newly diagnosed endometrial cancer who have undergone universal
12. Wang J, Xu P, Yang X, et al. Association of myometrial invasion with germline mutation testing. Cancer 2014;120:3932–3939.
lymphovascular space invasion, lymph node metastasis, recurrence, and 22. Goodfellow PJ, Billingsley CC, Lankes HA, et al. Combined microsatel-
overall survival in endometrial cancer: a meta-analysis of 79 studies with lite instability, MLH1 methylation analysis, and immunohistochemistry
68,870 patients. Front Oncol 2021;11:762329. for lynch syndrome screening in endometrial cancers from GOG210: an

202 © JNCCN—Journal of the National Comprehensive Cancer Network | Volume 21 Issue 2 | February 2023
Uterine Neoplasms, Version 1.2023 NCCN GUIDELINES®

NRG oncology and Gynecologic Oncology Group study. J Clin Oncol 47. Mariani A, Dowdy SC, Podratz KC. New surgical staging of endometrial
2015;33:4301–4308. cancer: 20 years later. Int J Gynaecol Obstet 2009;105:110–111.
23. Watkins JC, Yang EJ, Muto MG, et al. Universal screening for mismatch- 48. Amin MB, Edge SB, Greene FL, et al, eds. AJCC Cancer Staging
repair deficiency in endometrial cancers to identify patients with Lynch Manual. 8th ed. Springer; 2017.
syndrome and Lynch-like syndrome. Int J Gynecol Pathol 2017;36:115–127. 49. Matsuo K, Yabuno A, Hom MS, et al. Significance of abnormal perito-
24. Mills AM, Liou S, Ford JM, et al. Lynch syndrome screening should be neal cytology on survival of women with stage I-II endometrioid endo-
considered for all patients with newly diagnosed endometrial cancer. metrial cancer. Gynecol Oncol 2018;149:301–309.
Am J Surg Pathol 2014;38:1501–1509. 50. Peters EEM, Le on-Castillo A, Smit VT, et al. Defining substantial
25. Raffone A, Travaglino A, Cerbone M, et al. Diagnostic accuracy of lymphovascular space invasion in endometrial cancer. Int J Gynecol
immunohistochemistry for mismatch repair proteins as surrogate of Pathol 2022;41:220–226.
microsatellite instability molecular testing in endometrial cancer. Pathol
51. Brierley JD, Gospodarowicz MK, Wittekind C, eds. TNM Classification
Oncol Res 2020;26:1417–1427.
of Malignant Tumours. 8th ed. John Wiley & Sons; 2017.
26. Bonnet D, Selves J, Toulas C, et al. Simplified identification of Lynch
52. Rottmann D, Snir OL, Wu X, et al. HER2 testing of gynecologic carcino-
syndrome: a prospective, multicenter study. Dig Liver Dis 2012;44:
515–522. sarcomas: tumor stratification for potential targeted therapy. Mod
Pathol 2020;33:118–127.
27. Win AK, Lindor NM, Winship I, et al. Risks of colorectal and other can-
cers after endometrial cancer for women with Lynch syndrome. J Natl 53. Crane E, Naumann W, Tait D, et al. Molecular variations in uterine
Cancer Inst 2013;105:274–279. carcinosarcomas identify therapeutic opportunities. Int J Gynecol
Cancer 2020;30:480–484.
28. Manchanda R, Saridogan E, Abdelraheim A, et al. Annual outpatient
hysteroscopy and endometrial sampling (OHES) in HNPCC/Lynch 54. Livasy CA, Reading FC, Moore DT, et al. EGFR expression and HER2/
syndrome (LS). Arch Gynecol Obstet 2012;286:1555–1562. neu overexpression/amplification in endometrial carcinosarcoma.
Gynecol Oncol 2006;100:101–106.
29. Meyer LA, Broaddus RR, Lu KH. Endometrial cancer and Lynch
syndrome: clinical and pathologic considerations. Cancer Control 2009; 55. Yoshida H, Nishikawa T, Matsumoto K, et al. Histopathological features
16:14–22. of HER2 overexpression in uterine carcinosarcoma: proposal for require-
30. J€arvinen HJ, Renkonen-Sinisalo L, Akt an-Coll
an K, et al. Ten years after ments in HER2 testing for targeted therapy. Virchows Arch 2021;478:
mutation testing for Lynch syndrome: cancer incidence and outcome in 1161–1171.
mutation-positive and mutation-negative family members. J Clin Oncol 56. Jenkins TM, Cantrell LA, Stoler MH, et al. HER2 overexpression and
2009;27:4793–4797. amplification in uterine carcinosarcomas with serous morphology. Am J
31. Schmeler KM, Lynch HT, Chen LM, et al. Prophylactic surgery to reduce Surg Pathol 2022;46:435–442.
the risk of gynecologic cancers in the Lynch syndrome. N Engl J Med 57. Ali A, Black D, Soslow RA. Difficulties in assessing the depth of myome-
2006;354:261–269. trial invasion in endometrial carcinoma. Int J Gynecol Pathol 2007;26:
32. Leslie KK, Thiel KW, Goodheart MJ, et al. Endometrial cancer. Obstet 115–123.
Gynecol Clin North Am 2012;39:255–268. 58. Goff BA, Rice LW. Assessment of depth of myometrial invasion in endo-
33. Smith RA, Brooks D, Cokkinides V, et al. Cancer screening in the United metrial adenocarcinoma. Gynecol Oncol 1990;38:46–48.
States, 2013: a review of current American Cancer Society guidelines,
59. Daniel AG, Peters WA III. Accuracy of office and operating room curet-
current issues in cancer screening, and new guidance on cervical cancer
tage in the grading of endometrial carcinoma. Obstet Gynecol 1988;71:
screening and lung cancer screening. CA Cancer J Clin 2013;63:
612–614.
88–105.
60. Soliman PT, Frumovitz M, Spannuth W, et al. Lymphadenectomy during
34. McCluggage WG. My approach to the interpretation of endometrial
biopsies and curettings. J Clin Pathol 2006;59:801–812. endometrial cancer staging: practice patterns among gynecologic
oncologists. Gynecol Oncol 2010;119:291–294.
35. McKenney JK, Longacre TA. Low-grade endometrial adenocarcinoma: a
diagnostic algorithm for distinguishing atypical endometrial hyperplasia 61. Kumar S, Mariani A, Bakkum-Gamez JN, et al. Risk factors that mitigate
and other benign (and malignant) mimics. Adv Anat Pathol 2009;16: the role of paraaortic lymphadenectomy in uterine endometrioid cancer.
1–22. Gynecol Oncol 2013;130:441–445.
36. Leitao MM Jr, Kehoe S, Barakat RR, et al. Comparison of D&C and 62. Todo Y, Kato H, Kaneuchi M, et al. Survival effect of para-aortic lympha-
office endometrial biopsy accuracy in patients with FIGO grade 1 endo- denectomy in endometrial cancer (SEPAL study): a retrospective cohort
metrial adenocarcinoma. Gynecol Oncol 2009;113:105–108. analysis. Lancet 2010;375:1165–1172.
37. Crivellaro C, Signorelli M, Guerra L, et al. Tailoring systematic lympha- 63. Kitchener H, Swart AM, Qian Q, et al. Efficacy of systematic pelvic
denectomy in high-risk clinical early stage endometrial cancer: the role lymphadenectomy in endometrial cancer (MRC ASTEC trial): a rando-
of 18F-FDG PET/CT. Gynecol Oncol 2013;130:306–311. mised study. Lancet 2009;373:125–136.
38. Antonsen SL, Jensen LN, Loft A, et al. MRI, PET/CT and ultrasound in 64. Benedetti Panici P, Basile S, Maneschi F, et al. Systematic pelvic lympha-
the preoperative staging of endometrial cancer - a multicenter prospec- denectomy vs. no lymphadenectomy in early-stage endometrial carci-
tive comparative study. Gynecol Oncol 2013;128:300–308. noma: randomized clinical trial. J Natl Cancer Inst 2008;100:1707–1716.
39. Bollineni VR, Ytre-Hauge S, Bollineni-Balabay O, et al. High diagnostic 65. American College of Obstetricians and Gynecologists. ACOG practice
value of 18F-FDG PET/CT in endometrial cancer: systematic review and bulletin, clinical management guidelines for obstetrician-gynecologists,
meta-analysis of the literature. J Nucl Med 2016;57:879–885. number 65, August 2005: management of endometrial cancer. Obstet
40. Reinhold C, Ueno Y, Akin EA, et al. ACR Appropriateness Criteria Gynecol 2005;106:413–425.
pretreatment evaluation and follow-up of endometrial cancer. J Am Coll 66. Seamon LG, Fowler JM, Cohn DE. Lymphadenectomy for endometrial
Radiol 2020;17(Suppl 11):S472–486. cancer: the controversy. Gynecol Oncol 2010;117:6–8.
41. Patsner B, Orr JW Jr, Mann WJ Jr. Use of serum CA 125 measurement 67. Creasman WT, Mutch DE, Herzog TJ. ASTEC lymphadenectomy and
in posttreatment surveillance of early-stage endometrial carcinoma. Am radiation therapy studies: are conclusions valid? Gynecol Oncol 2010;
J Obstet Gynecol 1990;162:427–429. 116:293–294.
42. Rose PG, Sommers RM, Reale FR, et al. Serial serum CA 125 measure- 68. Uccella S, Podratz KC, Aletti GD, et al. Lymphadenectomy in endome-
ments for evaluation of recurrence in patients with endometrial carci- trial cancer. Lancet 2009;373:1170; author reply 1170–1171.
noma. Obstet Gynecol 1994;84:12–16.
69. Milam MR, Java J, Walker JL, et al. Nodal metastasis risk in endome-
43. Creasman WT, Morrow CP, Bundy BN, et al. Surgical pathologic spread
trioid endometrial cancer. Obstet Gynecol 2012;119:286–292.
patterns of endometrial cancer. A Gynecologic Oncology Group Study.
Cancer 1987;60(Suppl 8):2035–2041. 70. Neubauer NL, Lurain JR. The role of lymphadenectomy in surgical stag-
44. Benedet JL, Bender H, Jones H III, et al. FIGO staging classifications ing of endometrial cancer. Int J Surg Oncol 2011;2011:814649.
and clinical practice guidelines in the management of gynecologic 71. Bodurtha Smith AJ, Fader AN, Tanner EJ. Sentinel lymph node assess-
cancers. Int J Gynaecol Obstet 2000;70:209–262. ment in endometrial cancer: a systematic review and meta-analysis. Am
45. Wright JD, Barrena Medel NI, Sehouli J, et al. Contemporary manage- J Obstet Gynecol 2017;216:459–476.e10.
ment of endometrial cancer. Lancet 2012;379:1352–1360. 72. Mariani A, Dowdy SC, Cliby WA, et al. Prospective assessment of
46. Creasman W. Revised FIGO staging for carcinoma of the endometrium. lymphatic dissemination in endometrial cancer: a paradigm shift in
Int J Gynaecol Obstet 2009;105:109. surgical staging. Gynecol Oncol 2008;109:11–18.

JNCCN.org | Volume 21 Issue 2 | February 2023 203


NCCN GUIDELINES® Uterine Neoplasms, Version 1.2023

73. Hirahatake K, Hareyama H, Sakuragi N, et al. A clinical and pathologic 96. Holloway RW, Gupta S, Stavitzski NM, et al. Sentinel lymph node map-
study on para-aortic lymph node metastasis in endometrial carcinoma. ping with staging lymphadenectomy for patients with endometrial
J Surg Oncol 1997;65:82–87. cancer increases the detection of metastasis. Gynecol Oncol 2016;141:
74. Frederick PJ, Straughn JM Jr. The role of comprehensive surgical stag- 206–210.
ing in patients with endometrial cancer. Cancer Contr 2009;16:23–29. 97. Daraï E, Dubernard G, Bats AS, et al. Sentinel node biopsy for the
management of early stage endometrial cancer: long-term results of the
75. Barlin JN, Khoury-Collado F, Kim CH, et al. The importance of applying
SENTI-ENDO study. Gynecol Oncol 2015;136:54–59.
a sentinel lymph node mapping algorithm in endometrial cancer stag-
ing: beyond removal of blue nodes. Gynecol Oncol 2012;125:531–535. 98. Lee JH, Dubinsky T, Andreotti RF, et al. ACR Appropriateness Criteria
pretreatment evaluation and follow-up of endometrial cancer of the
76. Ballester M, Koskas M, Coutant C, et al. Does the use of the 2009 FIGO uterus. Ultrasound Q 2011;27:139–145.
classification of endometrial cancer impact on indications of the sentinel
99. Naoura I, Canlorbe G, Bendifallah S, et al. Relevance of sentinel lymph
node biopsy? BMC Cancer 2010;10:465.
node procedure for patients with high-risk endometrial cancer. Gynecol
77. How J, Lau S, Press J, et al. Accuracy of sentinel lymph node detection Oncol 2015;136:60–64.
following intra-operative cervical injection for endometrial cancer: 100. Schiavone MB, Zivanovic O, Zhou Q, et al. Survival of patients with
a prospective study. Gynecol Oncol 2012;127:332–337. uterine carcinosarcoma undergoing sentinel lymph node mapping.
78. Khoury-Collado F, Murray MP, Hensley ML, et al. Sentinel lymph node Ann Surg Oncol 2016;23:196–202.
mapping for endometrial cancer improves the detection of metastatic 101.  et al. Diagnostic accuracy and clinical
Lecointre L, Lodi M, Faller E,
disease to regional lymph nodes. Gynecol Oncol 2011;122:251–254. impact of sentinel lymph node sampling in endometrial cancer at high
79. Holloway RW, Abu-Rustum NR, Backes FJ, et al. Sentinel lymph node risk of recurrence: a meta-analysis. J Clin Med 2020;9:3874.
mapping and staging in endometrial cancer: a Society of Gynecologic 102. Bogani G, Papadia A, Buda A, et al. Sentinel node mapping vs sentinel
Oncology literature review with consensus recommendations. Gynecol node mapping plus back-up lymphadenectomy in high-risk endometrial
Oncol 2017;146:405–415. cancer patients: results from a multi-institutional study. Gynecol Oncol
80. Khoury-Collado F, St Clair C, Abu-Rustum NR. Sentinel lymph node 2021;161:122–129.
mapping in endometrial cancer: an update. Oncologist 2016;21: 103. Cusimano MC, Vicus D, Pulman K, et al. Assessment of sentinel lymph
461–466. node biopsy vs lymphadenectomy for intermediate- and high-grade
81. Rossi EC, Kowalski LD, Scalici J, et al. A comparison of sentinel lymph endometrial cancer staging. JAMA Surg 2021;156:157–164.
node biopsy to lymphadenectomy for endometrial cancer staging 104. Marchocki Z, Cusimano MC, Clarfield L, et al. Sentinel lymph node
(FIRES trial): a multicentre, prospective, cohort study. Lancet Oncol biopsy in high-grade endometrial cancer: a systematic review and meta-
2017;18:384–392. analysis of performance characteristics. Am J Obstet Gynecol 2021;225:
82. Paley PJ, Veljovich DS, Press JZ, et al. A prospective investigation of 367.E1–E39.
fluorescence imaging to detect sentinel lymph nodes at robotic-assisted 105. Euscher E, Sui D, Soliman P, et al. Ultrastaging of sentinel lymph nodes
endometrial cancer staging. Am J Obstet Gynecol 2016;215:117.E1–E7. in endometrial carcinoma according to use of 2 different methods. Int J
83. Ruscito I, Gasparri ML, Braicu EI, et al. Sentinel node mapping in cervi- Gynecol Pathol 2018;37:242–251.
cal and endometrial cancer: indocyanine green versus other conven- 106. Burg LC, Hengeveld EM, in ‘t Hout J, et al. Ultrastaging methods of
tional dyes-a meta-analysis. Ann Surg Oncol 2016;23:3749–3756. sentinel lymph nodes in endometrial cancer - a systematic review. Int J
Gynecol Cancer 2021;31:744–753.
84. Frumovitz M, Plante M, Lee PS, et al. Near-infrared fluorescence for
detection of sentinel lymph nodes in women with cervical and uterine 107. Kim CH, Soslow RA, Park KJ, et al. Pathologic ultrastaging improves
cancers (FILM): a randomised, phase 3, multicentre, non-inferiority trial. micrometastasis detection in sentinel lymph nodes during endometrial
Lancet Oncol 2018;19:1394–1403. cancer staging. Int J Gynecol Cancer 2013;23:964–970.
85. Ruiz R, Gorostidi M, Jaunarena I, et al. Maximizing sentinel node detec- 108. Raimond E, Ballester M, Hudry D, et al. Impact of sentinel lymph node
tion in endometrial cancer with dual cervical and transcervical fundal biopsy on the therapeutic management of early-stage endometrial
indocyanine green injection: 5-year single-center prospective study. Eur cancer: results of a retrospective multicenter study. Gynecol Oncol
J Obstet Gynecol Reprod Biol 2021;261:59–64. 2014;133:506–511.
86. Backes FJ, Cohen D, Salani R, et al. Prospective clinical trial of robotic 109. Amezcua CA, MacDonald HR, Lum CA, et al. Endometrial cancer
sentinel lymph node assessment with isosulfane blue (ISB) and indocya- patients have a significant risk of harboring isolated tumor cells in histolog-
ically negative lymph nodes. Int J Gynecol Cancer 2006;16:1336–1341.
nine green (ICG) in endometrial cancer and the impact of ultrastaging
(NCT01818739). Gynecol Oncol 2019;153:496–499. 110. Todo Y, Kato H, Okamoto K, et al. Isolated tumor cells and micrometa-
stases in regional lymph nodes in stage I to II endometrial cancer.
87. Vidal F, Leguevaque P, Motton S, et al. Evaluation of the sentinel lymph
J Gynecol Oncol 2016;27:e1.
node algorithm with blue dye labeling for early-stage endometrial can-
cer in a multicentric setting. Int J Gynecol Cancer 2013;23:1237–1243. 111. Frimer M, Khoury-Collado F, Murray MP, et al. Micrometastasis of endo-
metrial cancer to sentinel lymph nodes: is it an artifact of uterine manip-
88. Abu-Rustum NR, Khoury-Collado F, Pandit-Taskar N, et al. Sentinel
ulation? Gynecol Oncol 2010;119:496–499.
lymph node mapping for grade 1 endometrial cancer: is it the answer
to the surgical staging dilemma? Gynecol Oncol 2009;113:163–169. 112. Smerage JB, Barlow WE, Hortobagyi GN, et al. Circulating tumor cells
and response to chemotherapy in metastatic breast cancer: SWOG
89. Ballester M, Dubernard G, L ecuru F, et al. Detection rate and diagnostic S0500. J Clin Oncol 2014;32:3483–3489.
accuracy of sentinel-node biopsy in early stage endometrial cancer: a
prospective multicentre study (SENTI-ENDO). Lancet Oncol 2011;12: 113. Olawaiye AB, Mutch DG. Lymphnode staging update in the American
469–476. Joint Committee on Cancer 8th edition cancer staging manual. Gynecol
Oncol 2018;150:7–8.
90. Press JZ, Gotlieb WH. Controversies in the treatment of early stage en-
114. St Clair CM, Eriksson AG, Ducie JA, et al. Low-volume lymph node me-
dometrial carcinoma. Obstet Gynecol Int 2012;2012:578490.
tastasis discovered during sentinel lymph node mapping for endome-
91. Touhami O, Trinh XB, Gregoire J, et al. Predictors of non-sentinel lymph trial carcinoma. Ann Surg Oncol 2016;23:1653–1659.
node (non-SLN) metastasis in patients with sentinel lymph node (SLN)
115. Plante M, Stanleigh J, Renaud MC, et al. Isolated tumor cells identified
metastasis in endometrial cancer. Gynecol Oncol 2015;138:41–45.
by sentinel lymph node mapping in endometrial cancer: does adjuvant
92. Burke WM, Orr J, Leitao M, et al. Endometrial cancer: a review and cur- treatment matter? Gynecol Oncol 2017;146:240–246.
rent management strategies: part I. Gynecol Oncol 2014;134:385–392.
116. G omez-Hidalgo NR, Ramirez PT, Ngo B, et al. Oncologic impact of
93. Soliman PT, Westin SN, Dioun S, et al. A prospective validation study of micrometastases or isolated tumor cells in sentinel lymph nodes of
sentinel lymph node mapping for high-risk endometrial cancer. Gynecol patients with endometrial cancer: a meta-analysis. Clin Transl Oncol
Oncol 2017;146:234–239. 2020;22:1272–1279.
94. Kim CH, Khoury-Collado F, Barber EL, et al. Sentinel lymph node map- 117. Scalici J, Laughlin BB, Finan MA, et al. The trend towards minimally
ping with pathologic ultrastaging: a valuable tool for assessing nodal invasive surgery (MIS) for endometrial cancer: an ACS-NSQIP evaluation
metastasis in low-grade endometrial cancer with superficial myoinva- of surgical outcomes. Gynecol Oncol 2015;136:512–515.
sion. Gynecol Oncol 2013;131:714–719. 118. Janda M, Gebski V, Davies LC, et al. Effect of total laparoscopic hyster-
95. Cormier B, Rozenholc AT, Gotlieb W, et al. Sentinel lymph node proce- ectomy vs total abdominal hysterectomy on disease-free survival among
dure in endometrial cancer: a systematic review and proposal for stan- women with stage I endometrial cancer: a randomized clinical trial.
dardization of future research. Gynecol Oncol 2015;138:478–485. JAMA 2017;317:1224–1233.

204 © JNCCN—Journal of the National Comprehensive Cancer Network | Volume 21 Issue 2 | February 2023
Uterine Neoplasms, Version 1.2023 NCCN GUIDELINES®

119. Fader AN, Weise RM, Sinno AK, et al. Utilization of minimally invasive 142. Backes FJ, Brudie LA, Farrell MR, et al. Short- and long-term morbidity
surgery in endometrial cancer care: a quality and cost disparity. Obstet and outcomes after robotic surgery for comprehensive endometrial can-
Gynecol 2016;127:91–100. cer staging. Gynecol Oncol 2012;125:546–551.
120. Galaal K, Bryant A, Fisher AD, et al. Laparoscopy versus laparotomy for 143. Fleming ND, Ramirez PT. Robotic surgery in gynecologic oncology.
the management of early stage endometrial cancer. Cochrane Database Curr Opin Oncol 2012;24:547–553.
Syst Rev 2012;9:CD006655. 144. Siesto G, Ornaghi S, Ied a N, et al. Robotic surgical staging for endome-
121. Kornblith AB, Huang HQ, Walker JL, et al. Quality of life of patients trial and cervical cancers in medically ill patients. Gynecol Oncol 2013;
with endometrial cancer undergoing laparoscopic international federa- 129:593–597.
tion of gynecology and obstetrics staging compared with laparotomy: a 145. van Dam P, Hauspy J, Verkinderen L, et al. Are costs of robot-assisted
Gynecologic Oncology Group study. J Clin Oncol 2009;27:5337–5342. surgery warranted for gynecological procedures? Obstet Gynecol Int
122. Walker JL, Piedmonte MR, Spirtos NM, et al. Laparoscopy compared 2011;2011:973830.
with laparotomy for comprehensive surgical staging of uterine cancer: 146. Weinberg L, Rao S, Escobar PF. Robotic surgery in gynecology:
Gynecologic Oncology Group study LAP2. J Clin Oncol 2009;27: an updated systematic review. Obstet Gynecol Int 2011;2011:852061.
5331–5336. 147. Ramirez PT, Adams S, Boggess JF, et al. Robotic-assisted surgery in
123. Mannschreck D, Matsuno RK, Moriarty JP, et al. Disparities in surgical gynecologic oncology: a Society of Gynecologic Oncology consensus
care among women with endometrial cancer. Obstet Gynecol 2016; statement. Gynecol Oncol 2012;124:180–184.
128:526–534. 148. AAGL Advancing Minimally Invasive Gynecology Worldwide. Guidelines
124. Walker JL, Piedmonte MR, Spirtos NM, et al. Recurrence and survival for privileging for robotic-assisted gynecologic laparoscopy. J Minim In-
after random assignment to laparoscopy versus laparotomy for compre- vasive Gynecol 2014;21:157–167.
hensive surgical staging of uterine cancer: Gynecologic Oncology 149. American Congress of Obstetricians and Gynecologists. Statement on
Group LAP2 Study. J Clin Oncol 2012;30:695–700. Robotic Surgery by ACOG President James T. Breeden. Accessed
125. King LP, Miller DS. Recent progress: gynecologic oncology group trials December 20, 2022. Available at: http://www.acog.org/About-ACOG/
News-Room/News-Releases/2013/Statement-on-Robotic-Surgery
in uterine corpus tumors. Rev Recent Clin Trials 2009;4:70–74.
150. Sinno AK, Fader AN. Robotic-assisted surgery in gynecologic oncology.
126. Vergote I, Amant F, Neven P. Is it safe to treat endometrial carcinoma
Fertil Steril 2014;102:922–932.
endoscopically? J Clin Oncol 2009;27:5305–5307.
151. Gala RB, Margulies R, Steinberg A, et al. Systematic review of robotic
127. Mourits MJ, Bijen CB, Arts HJ, et al. Safety of laparoscopy versus lapa-
surgery in gynecology: robotic techniques compared with laparoscopy
rotomy in early-stage endometrial cancer: a randomised trial. Lancet
and laparotomy. J Minim Invasive Gynecol 2014;21:353–361.
Oncol 2010;11:763–771.
152. Society of Gynecologic Onconlogy. SGO position statement: morcella-
128. Reijntjes B, van Suijlichem M, Woolderink JM, et al. Recurrence and sur- tion. Accessed September 30, 2014. Available at: https://www.sgo.org/
vival after laparoscopy versus laparotomy without lymphadenectomy in newsroom/position-statements-2/morcellation/
early-stage endometrial cancer: long-term outcomes of a randomised
153. ACOG. Power morcellation and occult malignancy in gynecologic sur-
trial. Gynecol Oncol 2022;164:265–270.
gery. Accessed January 23, 2023. Available at: https://www.sgo.org/wp-
129. He H, Zeng D, Ou H, et al. Laparoscopic treatment of endometrial can- content/uploads/2014/04/ACOG_Statement.pdf
cer: systematic review. J Minim Invasive Gynecol 2013;20:413–423.
154. U.S. Department of Health and Human Services. FDA discourages use
130. Wang HL, Ren YF, Yang J, et al. Total laparoscopic hysterectomy versus of laparoscopic power morcellation for removal of uterus or uterine fib-
total abdominal hysterectomy for endometrial cancer: a meta-analysis. roids. Accessed January 23, 2023. Available at: http://www.fda.gov/
Asian Pac J Cancer Prev 2013;14:2515–2519. NewsEvents/Newsroom/PressAnnouncements/ucm393689.htm
131. Mori KM, Neubauer NL. Minimally invasive surgery in gynecologic on- 155. Bogani G, Cliby WA, Aletti GD. Impact of morcellation on survival
cology. ISRN Obstet Gynecol 2013;2013:312982. outcomes of patients with unexpected uterine leiomyosarcoma: a
132. Krill LS, Bristow RE. Robotic surgery: gynecologic oncology. Cancer J systematic review and meta-analysis. Gynecol Oncol 2015;137:
2013;19:167–176. 167–172.
133. ElSahwi KS, Hooper C, De Leon MC, et al. Comparison between 155 156. Wright JD, Buck AM, Shah M, et al. Safety of ovarian preservation in
cases of robotic vs. 150 cases of open surgical staging for endometrial premenopausal women with endometrial cancer. J Clin Oncol 2009;27:
cancer. Gynecol Oncol 2012;124:260–264. 1214–1219.
134. Chan JK, Gardner AB, Taylor K, et al. Robotic versus laparoscopic ver- 157. Koskas M, Bendifallah S, Luton D, et al. Safety of uterine and/or ovarian
sus open surgery in morbidly obese endometrial cancer patients - a preservation in young women with grade 1 intramucous endometrial
comparative analysis of total charges and complication rates. Gynecol adenocarcinoma: a comparison of survival according to the extent of
surgery. Fertil Steril 2012;98:1229–1235.
Oncol 2015;139:300–305.
158. Lee TS, Lee JY, Kim JW, et al. Outcomes of ovarian preservation in a
135. Coronado PJ, Herraiz MA, Magrina JF, et al. Comparison of periopera-
cohort of premenopausal women with early-stage endometrial cancer: a
tive outcomes and cost of robotic-assisted laparoscopy, laparoscopy
Korean Gynecologic Oncology Group study. Gynecol Oncol 2013;131:
and laparotomy for endometrial cancer. Eur J Obstet Gynecol Reprod
289–293.
Biol 2012;165:289–294.
159. Manfredi R, Mirk P, Maresca G, et al. Local-regional staging of endome-
136. Seamon LG, Cohn DE, Henretta MS, et al. Minimally invasive compre- trial carcinoma: role of MR imaging in surgical planning. Radiology
hensive surgical staging for endometrial cancer: robotics or laparos- 2004;231:372–378.
copy? Gynecol Oncol 2009;113:36–41.
160. Akin O, Mironov S, Pandit-Taskar N, et al. Imaging of uterine cancer.
137. Bell MC, Torgerson J, Seshadri-Kreaden U, et al. Comparison of out- Radiol Clin North Am 2007;45:167–182.
comes and cost for endometrial cancer staging via traditional laparot-
161. Gunderson CC, Fader AN, Carson KA, et al. Oncologic and reproduc-
omy, standard laparoscopy and robotic techniques. Gynecol Oncol
tive outcomes with progestin therapy in women with endometrial hyper-
2008;111:407–411.
plasia and grade 1 adenocarcinoma: a systematic review. Gynecol
138. Cardenas-Goicoechea J, Adams S, Bhat SB, et al. Surgical outcomes of Oncol 2012;125:477–482.
robotic-assisted surgical staging for endometrial cancer are equivalent 162. Baker J, Obermair A, Gebski V, et al. Efficacy of oral or intrauterine
to traditional laparoscopic staging at a minimally invasive surgical cen- device-delivered progestin in patients with complex endometrial hyperpla-
ter. Gynecol Oncol 2010;117:224–228. sia with atypia or early endometrial adenocarcinoma: a meta-analysis and
139. Abel MK, Chan JK, Chow S, et al. Trends and survival outcomes of systematic review of the literature. Gynecol Oncol 2012;125:263–270.
robotic, laparoscopic, and open surgery for stage II uterine cancer. Int J 163. Gracia CR, Jeruss JS. Lives in the balance: women with cancer and the
Gynecol Cancer 2020;30:1347–1355. right to fertility care. J Clin Oncol 2013;31:668–669.
140. Capozzi VA, Riemma G, Rosati A, et al. Surgical complications occurring 164. Ushijima K, Yahata H, Yoshikawa H, et al. Multicenter phase II study of
during minimally invasive sentinel lymph node detection in endometrial fertility-sparing treatment with medroxyprogesterone acetate for endo-
cancer patients. A systematic review of the literature and metanalysis. metrial carcinoma and atypical hyperplasia in young women. J Clin
Eur J Surg Oncol 2021;47:2142–2149. Oncol 2007;25:2798–2803.
141. Brudie LA, Backes FJ, Ahmad S, et al. Analysis of disease recurrence 165. Hubbs JL, Saig RM, Abaid LN, et al. Systemic and local hormone ther-
and survival for women with uterine malignancies undergoing robotic apy for endometrial hyperplasia and early adenocarcinoma. Obstet
surgery. Gynecol Oncol 2013;128:309–315. Gynecol 2013;121:1172–1180.

JNCCN.org | Volume 21 Issue 2 | February 2023 205


NCCN GUIDELINES® Uterine Neoplasms, Version 1.2023

166. Trimble CL, Method M, Leitao M, et al. Management of endometrial 189. Creutzberg CL, Nout RA. The role of radiotherapy in endometrial
precancers. Obstet Gynecol 2012;120:1160–1175. cancer: current evidence and trends. Curr Oncol Rep 2011;13:472–478.
167. Mehasseb MK, Latimer JA. Controversies in the management of endo- 190. Klopp A, Smith BD, Alektiar K, et al. The role of postoperative radiation
metrial carcinoma: an update. Obstet Gynecol Int 2012;2012:676032. therapy for endometrial cancer: executive summary of an American
168. Hahn HS, Yoon SG, Hong JS, et al. Conservative treatment with proges- Society for Radiation Oncology evidence-based guideline. Pract Radiat
tin and pregnancy outcomes in endometrial cancer. Int J Gynecol Can- Oncol 2014;4:137–144.
cer 2009;19:1068–1073. 191. Meyer LA, Bohlke K, Powell MA, et al. Postoperative radiation therapy
169. Park JY, Kim DY, Kim JH, et al. Long-term oncologic outcomes after fer- for endometrial cancer: American Society of Clinical Oncology clinical
tility-sparing management using oral progestin for young women with practice guideline endorsement of the American Society for Radiation
endometrial cancer (KGOG 2002). Eur J Cancer 2013;49:868–874. Oncology evidence-based guideline. J Clin Oncol 2015;33:2908–2913.
170. Bi Q, Bi G, Wang J, et al. Diagnostic accuracy of MRI for detecting 192. Neubauer NL, Havrilesky LJ, Calingaert B, et al. The role of lymphade-
cervical invasion in patients with endometrial carcinoma: a meta- nectomy in the management of preoperative grade 1 endometrial carci-
analysis. J Cancer 2021;12:754–764. noma. Gynecol Oncol 2009;112:511–516.
171. Bi Q, Chen Y, Wu K, et al. The diagnostic value of MRI for preoperative 193. Gretz HF III, Economos K, Husain A, et al. The practice of surgical stag-
staging in patients with endometrial cancer: a meta-analysis. Acad ing and its impact on adjuvant treatment recommendations in patients
Radiol 2020;27:960–968. with stage I endometrial carcinoma. Gynecol Oncol 1996;61:409–415.
172. Boente MP, Yordan EL Jr, McIntosh DG, et al. Prognostic factors and 194. Ben-Shachar I, Pavelka J, Cohn DE, et al. Surgical staging for patients
long-term survival in endometrial adenocarcinoma with cervical involve- presenting with grade 1 endometrial carcinoma. Obstet Gynecol 2005;
ment. Gynecol Oncol 1993;51:316–322. 105:487–493.
173. Sartori E, Gadducci A, Landoni F, et al. Clinical behavior of 203 stage II 195. Nout RA, Smit VT, Putter H, et al. Vaginal brachytherapy versus pelvic
endometrial cancer cases: the impact of primary surgical approach and external beam radiotherapy for patients with endometrial cancer of
of adjuvant radiation therapy. Int J Gynecol Cancer 2001;11:430–437. high-intermediate risk (PORTEC-2): an open-label, non-inferiority, rando-
174. Barlin JN, Puri I, Bristow RE. Cytoreductive surgery for advanced or mised trial. Lancet 2010;375:816–823.
recurrent endometrial cancer: a meta-analysis. Gynecol Oncol 2010; 196. Elshaikh MA, Al-Wahab Z, Mahdi H, et al. Recurrence patterns and sur-
118:14–18. vival endpoints in women with stage II uterine endometrioid carcinoma:
175. Lambrou NC, G omez-Marın O, Mirhashemi R, et al. Optimal surgical cy- a multi-institution study. Gynecol Oncol 2015;136:235–239.
toreduction in patients with stage III and stage IV endometrial carcinoma: 197. Creutzberg CL, van Putten WL, Koper PC, et al. Surgery and postopera-
a study of morbidity and survival. Gynecol Oncol 2004;93:653–658. tive radiotherapy versus surgery alone for patients with stage-1 endo-
176. Albright BB, Monuszko KA, Kaplan SJ, et al. Primary cytoreductive sur- metrial carcinoma: multicentre randomised trial. Lancet 2000;355:
gery for advanced stage endometrial cancer: a systematic review and 1404–1411.
meta-analysis. Am J Obstet Gynecol 2021;225:237.e1–237.e24. 198. Aalders J, Abeler V, Kolstad P, et al. Postoperative external irradiation
177. Palma DA, Olson R, Harrow S, et al. Stereotactic ablative radiotherapy and prognostic parameters in stage I endometrial carcinoma: clinical
versus standard of care palliative treatment in patients with oligometa- and histopathologic study of 540 patients. Obstet Gynecol 1980;56:
static cancers (SABR-COMET): a randomised, phase 2, open-label trial. 419–427.
Lancet 2019;393:2051–2058. 199. Keys HM, Roberts JA, Brunetto VL, et al. A phase III trial of surgery with
178. Gadducci A, Cosio S, Genazzani AR. Old and new perspectives in the or without adjunctive external pelvic radiation therapy in intermediate
pharmacological treatment of advanced or recurrent endometrial can- risk endometrial adenocarcinoma: a Gynecologic Oncology Group
cer: hormonal therapy, chemotherapy and molecularly targeted thera- study. Gynecol Oncol 2004;92:744–751.
pies. Crit Rev Oncol Hematol 2006;58:242–256. 200. Creutzberg CL, van Putten WL, W arl
am-Rodenhuis CC, et al. Outcome
179. Fishman DA, Roberts KB, Chambers JT, et al. Radiation therapy as ex- of high-risk stage IC, grade 3, compared with stage I endometrial carci-
clusive treatment for medically inoperable patients with stage I and II noma patients: the Postoperative Radiation Therapy in Endometrial
endometrioid carcinoma with endometrium. Gynecol Oncol 1996;61: Carcinoma Trial. J Clin Oncol 2004;22:1234–1241.
189–196.
201. Scholten AN, van Putten WL, Beerman H, et al. Postoperative radiother-
180. Coon D, Beriwal S, Heron DE, et al. High-dose-rate Rotte “Y” applicator apy for stage 1 endometrial carcinoma: long-term outcome of the ran-
brachytherapy for definitive treatment of medically inoperable endome- domized PORTEC trial with central pathology review. Int J Radiat Oncol
trial cancer: 10-year results. Int J Radiat Oncol Biol Phys 2008;71: Biol Phys 2005;63:834–838.
779–783.
202. Blake P, Swart AM, Orton J, et al. Adjuvant external beam radiotherapy
181. Niazi TM, Souhami L, Portelance L, et al. Long-term results of high- in the treatment of endometrial cancer (MRC ASTEC and NCIC CTG
dose-rate brachytherapy in the primary treatment of medically inoperable EN.5 randomised trials): pooled trial results, systematic review, and
stage I-II endometrial carcinoma. Int J Radiat Oncol Biol Phys 2005;63: meta-analysis. Lancet 2009;373:137–146.
1108–1113.
203. H€ockel M, Dornh€ ofer N. Treatment of early endometrial carcinoma: is
182. van der Steen-Banasik E, Christiaens M, Shash E, et al. Systemic review: less more? Lancet 2009;373:97–99.
radiation therapy alone in medical non-operable endometrial carci-
noma. Eur J Cancer 2016;65:172–181. 204. Creutzberg CL, Nout RA, Lybeert ML, et al. Fifteen-year radiotherapy
outcomes of the randomized PORTEC-1 trial for endometrial carcinoma.
183. Smith SC, Koh WJ. Palliative radiation therapy for gynaecological malig-
Int J Radiat Oncol Biol Phys 2011;81:e631–638.
nancies. Best Pract Res Clin Obstet Gynaecol 2001;15:265–278.
205. Alektiar KM, Venkatraman E, Chi DS, et al. Intravaginal brachytherapy
184. Randall ME, Filiaci VL, Muss H, et al. Randomized phase III trial of
alone for intermediate-risk endometrial cancer. Int J Radiat Oncol Biol
whole-abdominal irradiation versus doxorubicin and cisplatin chemo-
Phys 2005;62:111–117.
therapy in advanced endometrial carcinoma: a Gynecologic Oncology
Group study. J Clin Oncol 2006;24:36–44. 206. Small W Jr, Beriwal S, Demanes DJ, et al. American Brachytherapy
Society consensus guidelines for adjuvant vaginal cuff brachytherapy
185. Homesley HD, Filiaci V, Gibbons SK, et al. A randomized phase III trial
after hysterectomy. Brachytherapy 2012;11:58–67.
in advanced endometrial carcinoma of surgery and volume directed
radiation followed by cisplatin and doxorubicin with or without pacli- 207. Onsrud M, Cvancarova M, Hellebust TP, et al. Long-term outcomes
taxel: a Gynecologic Oncology Group study. Gynecol Oncol 2009;112: after pelvic radiation for early-stage endometrial cancer. J Clin Oncol
543–552. 2013;31:3951–3956.
186. Secord AA, Havrilesky LJ, O’Malley DM, et al. A multicenter evaluation 208. Nout RA, Putter H, J€ urgenliemk-Schulz IM, et al. Five-year quality of life
of sequential multimodality therapy and clinical outcome for the treat- of endometrial cancer patients treated in the randomised Post Opera-
ment of advanced endometrial cancer. Gynecol Oncol 2009;114: tive Radiation Therapy in Endometrial Cancer (PORTEC-2) trial and com-
442–447. parison with norm data. Eur J Cancer 2012;48:1638–1648.
187. Creutzberg CL, van Stiphout RG, Nout RA, et al. Nomograms for 209. R€oper B, Astner ST, Heydemann-Obradovic A, et al. Ten-year data on
prediction of outcome with or without adjuvant radiation therapy for 138 patients with endometrial carcinoma and postoperative vaginal
patients with endometrial cancer: a pooled analysis of PORTEC-1 and brachytherapy alone: no need for external-beam radiotherapy in low
PORTEC-2 trials. Int J Radiat Oncol Biol Phys 2015;91:530–539. and intermediate risk patients. Gynecol Oncol 2007;107:541–548.
188. Burke WM, Orr J, Leitao M, et al. Endometrial cancer: a review and cur- 210. Nout RA, Putter H, J€ urgenliemk-Schulz IM, et al. Quality of life after
rent management strategies: part II. Gynecol Oncol 2014;134:393–402. pelvic radiotherapy or vaginal brachytherapy for endometrial cancer:

206 © JNCCN—Journal of the National Comprehensive Cancer Network | Volume 21 Issue 2 | February 2023
Uterine Neoplasms, Version 1.2023 NCCN GUIDELINES®

first results of the randomized PORTEC-2 trial. J Clin Oncol 2009;27: stage III, IV, and recurrent endometrial cancer. Gynecol Oncol 2011;
3547–3556. 121:112–117.
211. McCloskey SA, Tchabo NE, Malhotra HK, et al. Adjuvant vaginal brachy- 234. Brown AP, Gaffney DK, Dodson MK, et al. Survival analysis of endome-
therapy alone for high risk localized endometrial cancer as defined by trial cancer patients with positive lymph nodes. Int J Gynecol Cancer
the three major randomized trials of adjuvant pelvic radiation. Gynecol 2013;23:861–868.
Oncol 2010;116:404–407. 235. Lum MM, Belnap TW, Frandsen J, et al. Survival analysis of cancer
212. Dunn EF, Geye H, Platta CS, et al. Predictive factors of recurrence fol- patients with FIGO stage IIIA endometrial cancer. Am J Clin Oncol
lowing adjuvant vaginal cuff brachytherapy alone for stage I endometrial 2015;38:283–288.
cancer. Gynecol Oncol 2014;133:494–498. 236. Ren Y, Huang X, Shan B, et al. Adjuvant concurrent chemoradiation
213. Narasimhulu DM, Cope A, Riaz IB, et al. External beam radiotherapy followed by chemotherapy for high-risk endometrial cancer. Gynecol
versus vaginal brachytherapy in patients with stage II endometrial Oncol 2016;140:58–63.
cancer: a systematic review and meta-analysis. Int J Gynecol Cancer 237. Greven K, Winter K, Underhill K, et al. Final analysis of RTOG 9708:
2020;30:797–805. adjuvant postoperative irradiation combined with cisplatin/paclitaxel
214. Randall ME, Filiaci V, McMeekin DS, et al. Phase III trial: adjuvant pelvic chemotherapy following surgery for patients with high-risk endometrial
radiation therapy versus vaginal brachytherapy plus paclitaxel/carbopla- cancer. Gynecol Oncol 2006;103:155–159.
tin in high-intermediate and high-risk early stage endometrial cancer. 238. de Boer SM, Powell ME, Mileshkin L, et al. Adjuvant chemoradiotherapy
J Clin Oncol 2019;37:1810–1818. versus radiotherapy alone in women with high-risk endometrial cancer
215. Chino JP, Jones E, Berchuck A, et al. The influence of radiation modal- (PORTEC-3): patterns of recurrence and post-hoc survival analysis of a
ity and lymph node dissection on survival in early-stage endometrial randomised phase 3 trial. Lancet Oncol 2019;20:1273–1285.
cancer. Int J Radiat Oncol Biol Phys 2012;82:1872–1879. 239. Post CCB, de Boer SM, Powell ME, et al. Long-term toxicity and health-
216. Lee CM, Szabo A, Shrieve DC, et al. Frequency and effect of adjuvant related quality of life after adjuvant chemoradiation therapy or radiation
radiation therapy among women with stage I endometrial adenocarci- therapy alone for high-risk endometrial cancer in the randomized
noma. JAMA 2006;295:389–397. PORTEC-3 trial. Int J Radiat Oncol Biol Phys 2021;109:975–986.
217. Johnson N, Cornes P. Survival and recurrent disease after postoperative 240. Matei D, Filiaci V, Randall ME, et al. Adjuvant chemotherapy plus radiation for
radiotherapy for early endometrial cancer: systematic review and meta- locally advanced endometrial cancer. N Engl J Med 2019;380:2317–2326.
analysis. BJOG 2007;114:1313–1320. 241. Leon-Castillo A, de Boer SM, Powell ME, et al. Molecular classification of
218. Kong A, Johnson N, Cornes P, et al. Adjuvant radiotherapy for stage I the PORTEC-3 trial for high-risk endometrial cancer: impact on prognosis
endometrial cancer. Cochrane Database Syst Rev 2007;2:CD003916. and benefit from adjuvant therapy. J Clin Oncol 2020;38:3388–3397.
219. Kong A, Johnson N, Kitchener HC, et al. Adjuvant radiotherapy for 242. Wortman BG, Bosse T, Nout RA, et al. Molecular-integrated risk profile
stage I endometrial cancer: an updated Cochrane systematic review to determine adjuvant radiotherapy in endometrial cancer: evaluation of
and meta-analysis. J Natl Cancer Inst 2012;104:1625–1634. the pilot phase of the PORTEC-4a trial. Gynecol Oncol 2018;151:69–75.
220. Eifel PJ. The role of adjuvant radiation therapy for stage I endometrial 243. Barakat RR, Goldman NA, Patel DA, et al. Pelvic exenteration for recur-
cancer: does meta-analysis reveal the answer? J Natl Cancer Inst 2012; rent endometrial cancer. Gynecol Oncol 1999;75:99–102.
104:1615–1616. 244. Fleisch MC, Pantke P, Beckmann MW, et al. Predictors for long-term
221. Park HJ, Nam EJ, Kim S, et al. The benefit of adjuvant chemotherapy survival after interdisciplinary salvage surgery for advanced or recurrent
combined with postoperative radiotherapy for endometrial cancer: a gynecologic cancers. J Surg Oncol 2007;95:476–484.
meta-analysis. Eur J Obstet Gynecol Reprod Biol 2013;170:39–44. 245. Dowdy SC, Mariani A, Cliby WA, et al. Radical pelvic resection and
222. Hogberg T, Signorelli M, de Oliveira CF, et al. Sequential adjuvant intraoperative radiation therapy for recurrent endometrial cancer: tech-
chemotherapy and radiotherapy in endometrial cancer–results from two nique and analysis of outcomes. Gynecol Oncol 2006;101:280–286.
randomised studies. Eur J Cancer 2010;46:2422–2431. 246. Tran PT, Su Z, Hara W, et al. Long-term survivors using intraoperative
223. Johnson N, Bryant A, Miles T, et al. Adjuvant chemotherapy for endo- radiotherapy for recurrent gynecologic malignancies. Int J Radiat Oncol
metrial cancer after hysterectomy. Cochrane Database Syst Rev 2011; Biol Phys 2007;69:504–511.
2011:CD003175. 247. Creutzberg CL, van Putten WL, Koper PC, et al. Survival after relapse in
224. Hogberg T. Adjuvant chemotherapy in endometrial carcinoma: overview patients with endometrial cancer: results from a randomized trial. Gyne-
of randomised trials. Clin Oncol (R Coll Radiol) 2008;20:463–469. col Oncol 2003;89:201–209.
225. Secord AA, Geller MA, Broadwater G, et al. A multicenter evaluation of 248. Lin LL, Grigsby PW, Powell MA, et al. Definitive radiotherapy in the
adjuvant therapy in women with optimally resected stage IIIC endome- management of isolated vaginal recurrences of endometrial cancer. Int
trial cancer. Gynecol Oncol 2013;128:65–70. J Radiat Oncol Biol Phys 2005;63:500–504.
226. Greven KM, Lanciano RM, Corn B, et al. Pathologic stage III endome- 249. Altman AD, Thompson J, Nelson G, et al. Use of aromatase inhibitors
trial carcinoma. Prognostic factors and patterns of recurrence. Cancer as first- and second-line medical therapy in patients with endometrial
1993;71:3697–3702. adenocarcinoma: a retrospective study. J Obstet Gynaecol Can 2012;
227. Mustea A, Koensgen D, Belau A, et al. Adjuvant sequential chemoradia- 34:664–672.
tion therapy in high-risk endometrial cancer: results of a prospective, 250. Barker LC, Brand IR, Crawford SM. Sustained effect of the aromatase
multicenter phase-II study of the NOGGO (North-Eastern German Soci- inhibitors anastrozole and letrozole on endometrial thickness in patients
ety of Gynaecological Oncology). Cancer Chemother Pharmacol 2013; with endometrial hyperplasia and endometrial carcinoma. Curr Med Res
72:975–983. Opin 2009;25:1105–1109.
228. Jutzi L, Hoskins P, Lim P, et al. The importance of adjuvant chemotherapy 251. Fiorica JV, Brunetto VL, Hanjani P, et al. Phase II trial of alternating courses
and pelvic radiotherapy in high-risk early stage endometrial carcinoma. of megestrol acetate and tamoxifen in advanced endometrial carcinoma:
Gynecol Oncol 2013;131:581–585. a Gynecologic Oncology Group study. Gynecol Oncol 2004;92:10–14.
229. de Boer SM, Powell ME, Mileshkin L, et al. Toxicity and quality of life 252. Whitney CW, Brunetto VL, Zaino RJ, et al. Phase II study of medroxypro-
after adjuvant chemoradiotherapy versus radiotherapy alone for women gesterone acetate plus tamoxifen in advanced endometrial carcinoma: a
with high-risk endometrial cancer (PORTEC-3): an open-label, multi- Gynecologic Oncology Group study. Gynecol Oncol 2004;92:4–9.
centre, randomised, phase 3 trial. Lancet Oncol 2016;17:1114–1126. 253. Herzog TJ. What is the clinical value of adding tamoxifen to progestins
230. Gibbons S, Martinez A, Schray M, et al. Adjuvant whole abdominopelvic in the treatment [correction for treament] of advanced or recurrent
irradiation for high risk endometrial carcinoma. Int J Radiat Oncol Biol endometrial cancer? Gynecol Oncol 2004;92:1–3.
Phys 1991;21:1019–1025. 254. Singh M, Zaino RJ, Filiaci VJ, et al. Relationship of estrogen and proges-
231. Greer BE, Hamberger AD. Treatment of intraperitoneal metastatic terone receptors to clinical outcome in metastatic endometrial carcinoma:
adenocarcinoma of the endometrium by the whole-abdomen moving- a Gynecologic Oncology Group study. Gynecol Oncol 2007;106:325–333.
strip technique and pelvic boost irradiation. Gynecol Oncol 1983;16: 255. Kauppila A. Oestrogen and progestin receptors as prognostic indicators
365–373. in endometrial cancer. A review of the literature. Acta Oncol 1989;28:
232. Abaid LN, Rettenmaier MA, Brown JV III, et al. Sequential chemother- 561–566.
apy and radiotherapy as sandwich therapy for the treatment of high risk 256. Thigpen JT, Brady MF, Alvarez RD, et al. Oral medroxyprogesterone
endometrial cancer. J Gynecol Oncol 2012;23:22–27. acetate in the treatment of advanced or recurrent endometrial carci-
233. Geller MA, Ivy JJ, Ghebre R, et al. A phase II trial of carboplatin and noma: a dose-response study by the Gynecologic Oncology Group.
docetaxel followed by radiotherapy given in a “Sandwich” method for J Clin Oncol 1999;17:1736–1744.

JNCCN.org | Volume 21 Issue 2 | February 2023 207


NCCN GUIDELINES® Uterine Neoplasms, Version 1.2023

257. Decruze SB, Green JA. Hormone therapy in advanced and recurrent endo- 279. Traina TA, Sabbatini P, Aghajanian C, et al. Weekly topotecan for recur-
metrial cancer: a systematic review. Int J Gynecol Cancer 2007;17:964–978. rent endometrial cancer: a case series and review of the literature.
258. Dellinger TH, Monk BJ. Systemic therapy for recurrent endometrial can- Gynecol Oncol 2004;95:235–241.
cer: a review of North American trials. Expert Rev Anticancer Ther 2009; 280. Miller DS, Blessing JA, Lentz SS, et al. A phase II trial of topotecan in pa-
9:905–916. tients with advanced, persistent, or recurrent endometrial carcinoma: a
259. Thigpen T, Brady MF, Homesley HD, et al. Tamoxifen in the treatment Gynecologic Oncology Group study. Gynecol Oncol 2002;87:247–251.
of advanced or recurrent endometrial carcinoma: a Gynecologic Oncol- 281. Moxley KM, McMeekin DS. Endometrial carcinoma: a review of chemo-
ogy Group study. J Clin Oncol 2001;19:364–367. therapy, drug resistance, and the search for new agents. Oncologist
260. Pandya KJ, Yeap BY, Weiner LM, et al. Megestrol and tamoxifen in 2010;15:1026–1033.
patients with advanced endometrial cancer: an Eastern Cooperative 282. Muggia FM, Blessing JA, Sorosky J, et al. Phase II trial of the pegylated
Oncology Group study (E4882). Am J Clin Oncol 2001;24:43–46. liposomal doxorubicin in previously treated metastatic endometrial cancer:
261. Slomovitz BM, Jiang Y, Yates MS, et al. Phase II study of everolimus a Gynecologic Oncology Group study. J Clin Oncol 2002;20:2360–2364.
and letrozole in patients with recurrent endometrial carcinoma. J Clin 283. Garcia AA, Blessing JA, Nolte S, et al. A phase II evaluation of weekly
Oncol 2015;33:930–936. docetaxel in the treatment of recurrent or persistent endometrial carci-
262. Slomovitz BM, Filiaci VL, Walker JL, et al. A randomized phase II trial of noma: a study by the Gynecologic Oncology Group. Gynecol Oncol
everolimus and letrozole or hormonal therapy in women with advanced, 2008;111:22–26.
persistent or recurrent endometrial carcinoma: a GOG Foundation 284. Mountzios G, Pectasides D, Bournakis E, et al. Developments in the sys-
study. Gynecol Oncol 2022;164:481–491. temic treatment of endometrial cancer. Crit Rev Oncol Hematol 2011;
263. Quinn MA. Hormonal treatment of endometrial cancer. Hematol Oncol 79:278–292.
Clin North Am 1999;13:163–187; ix. 285. Aghajanian C, Sill MW, Darcy KM, et al. Phase II trial of bevacizumab in
264. Ray M, Fleming G. Management of advanced-stage and recurrent recurrent or persistent endometrial cancer: a Gynecologic Oncology
endometrial cancer. Semin Oncol 2009;36:145–154. Group study. J Clin Oncol 2011;29:2259–2265.
265. Humber CE, Tierney JF, Symonds RP, et al. Chemotherapy for 286. Ott PA, Bang YJ, Berton-Rigaud D, et al. Safety and antitumor activity
advanced, recurrent or metastatic endometrial cancer: a systematic of pembrolizumab in advanced programmed death ligand 1-positive
review of Cochrane collaboration. Ann Oncol 2007;18:409–420. endometrial cancer: results from the Keynote-028 study. J Clin Oncol
2017;35:2535–2541.
266. Cella D, Huang H, Homesley HD, et al. Patient-reported peripheral
neuropathy of doxorubicin and cisplatin with and without paclitaxel in 287. Le DT, Durham JN, Smith KN, et al. Mismatch repair deficiency predicts
the treatment of advanced endometrial cancer: results from GOG 184. response of solid tumors to PD-1 blockade. Science 2017;357:409–413.
Gynecol Oncol 2010;119:538–542. 288. Le DT, Uram JN, Wang H, et al. PD-1 blockade in tumors with mis-
267. Fleming GF, Brunetto VL, Cella D, et al. Phase III trial of doxorubicin match-repair deficiency. N Engl J Med 2015;372:2509–2520.
plus cisplatin with or without paclitaxel plus filgrastim in advanced 289. Prescribing information for pembrolizumab injection, for intravenous
endometrial carcinoma: a Gynecologic Oncology Group study. J Clin use. 2022. Accessed Jul 25, 2017. Available at: http://bit.ly/2cTmItE
Oncol 2004;22:2159–2166. 290. O’Malley DM, Bariani GM, Cassier PA, et al. Pembrolizumab in patients
268. Spirtos NM, Enserro D, Homesley HD, et al. The addition of paclitaxel with microsatellite instability-high advanced endometrial cancer: results
to doxorubicin and cisplatin and volume-directed radiation does not from the Keynote-158 study. J Clin Oncol 2022;40:752–761.
improve overall survival (OS) or long-term recurrence-free survival (RFS) 291. Subbiah V, Solit DB, Chan TA, et al. The FDA approval of pembrolizumab
in advanced endometrial cancer (EC): a randomized phase III NRG/ for adult and pediatric patients with tumor mutational burden (TMB) $10:
Gynecologic Oncology Group (GOG) study. Gynecol Oncol 2019;154: a decision centered on empowering patients and their physicians. Ann
13–21. Oncol 2020;31:1115–1118.
269. Sovak MA, Dupont J, Hensley ML, et al. Paclitaxel and carboplatin in 292. Bonneville R, Krook MA, Kautto EA, et al. Landscape of microsatellite insta-
the treatment of advanced or recurrent endometrial cancer: a large bility across 39 cancer types. JCO Precis Oncol 2017;2017:PO.17.00073.
retrospective study. Int J Gynecol Cancer 2007;17:197–203. 293. Prendergast EN, Holman LL, Liu AY, et al. Comprehensive genomic
270. Pectasides D, Xiros N, Papaxoinis G, et al. Carboplatin and paclitaxel in profiling of recurrent endometrial cancer: implications for selection of
advanced or metastatic endometrial cancer. Gynecol Oncol 2008;109: systemic therapy. Gynecol Oncol 2019;154:461–466.
250–254. 294. Makker V, Taylor MH, Aghajanian C, et al. Lenvatinib plus pembrolizumab
271. Sorbe B, Andersson H, Boman K, et al. Treatment of primary advanced in patients with advanced endometrial cancer. J Clin Oncol 2020;38:
and recurrent endometrial carcinoma with a combination of carboplatin 2981–2992.
and paclitaxel-long-term follow-up. Int J Gynecol Cancer 2008;18: 295. Makker V, Colombo N, Casado Herr aez A, et al. Lenvatinib plus pem-
803–808. brolizumab for advanced endometrial cancer. N Engl J Med 2022;386:
272. Miller DS, Filiaci VL, Mannel RS, et al. Carboplatin and paclitaxel for 437–448.
advanced endometrial cancer: final overall survival and adverse event 296. Azad NS, Gray RJ, Overman MJ, et al. Nivolumab is effective in
analysis of a phase III trial (NRG oncology/GOG0209). J Clin Oncol mismatch repair-deficient noncolorectal cancers: results from arm
2020;38:3841–3850. Z1D-A subprotocol of the NCI-MATCH (EAY131) study. J Clin Oncol
273. Nomura H, Aoki D, Takahashi F, et al. Randomized phase II study com- 2020;38:214–222.
paring docetaxel plus cisplatin, docetaxel plus carboplatin, and pacli- 297. Fader AN, Roque DM, Siegel E, et al. Randomized phase II trial of
taxel plus carboplatin in patients with advanced or recurrent carboplatin-paclitaxel versus carboplatin-paclitaxel-trastuzumab in
endometrial carcinoma: a Japanese Gynecologic Oncology Group study uterine serous carcinomas that overexpress human epidermal growth
(JGOG2041). Ann Oncol 2011;22:636–642. factor receptor 2/neu. J Clin Oncol 2018;36:2044–2051.
274. Simpkins F, Drake R, Escobar PF, et al. A phase II trial of paclitaxel, 298. Tymon-Rosario J, Siegel ER, Bellone S, et al. Trastuzumab tolerabil-
carboplatin, and bevacizumab in advanced and recurrent endometrial ity in the treatment of advanced (stage III-IV) or recurrent uterine
carcinoma (EMCA). Gynecol Oncol 2015;136:240–245. serous carcinomas that overexpress HER2/neu. Gynecol Oncol 2021;
275. Rose PG, Ali S, Moslemi-Kebria M, et al. Paclitaxel, carboplatin, and 163:93–99.
bevacizumab in advanced and recurrent endometrial carcinoma. Int J 299. Homesley HD, Filiaci V, Markman M, et al. Phase III trial of ifosfamide with
Gynecol Cancer 2017;27:452–458. or without paclitaxel in advanced uterine carcinosarcoma: a Gynecologic
276. Lorusso D, Ferrandina G, Colombo N, et al. Carboplatin-paclitaxel com- Oncology Group study. J Clin Oncol 2007;25:526–531.
pared to carboplatin-paclitaxel-bevacizumab in advanced or recurrent 300. Sutton G, Brunetto VL, Kilgore L, et al. A phase III trial of ifosfamide
endometrial cancer: MITO END-2 - a randomized phase II trial. Gynecol with or without cisplatin in carcinosarcoma of the uterus: a Gynecologic
Oncol 2019;155:406–412. Oncology Group study. Gynecol Oncol 2000;79:147–153.
277. Chen H, Liang M, Min J. Efficacy and safety of bevacizumab-combined 301. Sutton G, Kauderer J, Carson LF, et al. Adjuvant ifosfamide and cisplatin in
chemotherapy for advanced and recurrent endometrial cancer: patients with completely resected stage I or II carcinosarcomas (mixed me-
a systematic review and meta-analysis. Balkan Med J 2021;38:7–12. sodermal tumors) of the uterus: a Gynecologic Oncology Group study.
278. Wadler S, Levy DE, Lincoln ST, et al. Topotecan is an active agent in Gynecol Oncol 2005;96:630–634.
the first-line treatment of metastatic or recurrent endometrial carcinoma: 302. Galaal K, van der Heijden E, Godfrey K, et al. Adjuvant radiotherapy
Eastern Cooperative Oncology Group study E3E93. J Clin Oncol 2003; and/or chemotherapy after surgery for uterine carcinosarcoma.
21:2110–2114. Cochrane Database Syst Rev 2013;2013:CD006812.

208 © JNCCN—Journal of the National Comprehensive Cancer Network | Volume 21 Issue 2 | February 2023
Uterine Neoplasms, Version 1.2023 NCCN GUIDELINES®

Individual Disclosures for the NCCN Uterine Neoplasms Panel


Clinical Research Support/Data Scientific Advisory Boards, Promotional Advisory Boards,
Panel Member Safety Monitoring Board Consultant, or Expert Witness Consultant, or Speakers Bureau Specialties

Nadeem Abu-Rustum, MD GRAIL None Klinik Essen Germany Gynecology/Gynecologic oncology

Rebecca Arend, MD Champions Oncology, Inc.; Exelixis GSK; Kiyatec; Merck & Co., Inc.; Natera Gynecology/Gynecologic oncology
Inc.; GlaxoSmithKline; ImmunoGen, Seagen Inc.; SUTRO Biopharma,
Inc.; Merck & Co., Inc.; Plexxikon; Inc.; and VBL Therapeutics
and VBL Therapeutics

Emma Barber, MD Eli Lilly and Company Merck & Co., Inc. None Gynecology/Gynecologic oncology

Kristin Bradley, MDa None None None Radiation oncology

Rebecca Brooks, MD None AstraZeneca Pharmaceuticals LP; AstraZeneca Pharmaceuticals LP; Gynecology/Gynecologic oncology
GlaxoSmithKline; and O’Neill GlaxoSmithKline
McFadden & Willett LLP

Susana M. Campos, MD, MPH, MS None Agenus; AstraZeneca None Medical oncology
Pharmaceuticals LP; Eisai Inc.;
GlaxoSmithKline; Merck & Co., Inc.;
and Novartis Pharmaceuticals
Corporation

Junzo Chino, MD None None None Radiation oncology

Hye Sook Chon, MD None Eisai Inc. None Gynecology/Gynecologic oncology

Christina Chu, MD University of Pennsylvania Hura Imaging None Gynecology/Gynecologic oncology

Marta Ann Crispens, MD AstraZeneca Pharmaceuticals LP GlaxoSmithKline None Gynecology/Gynecologic oncology

Shari Damast, MD None None None Radiation oncology

Christine M. Fisher, MD, MPH Axio Biostatistics, LLC None None Radiation oncology

Peter Frederick, MD None None None Gynecology/Gynecologic oncology

David K. Gaffney, MD, PhD Elekta AstraZeneca Pharmaceuticals LP; None Radiation oncology
and Merck & Co., Inc.

Robert Giuntoli II, MD AstraZeneca Pharmaceuticals LP McCumber, Daniels, Buntz, Hartig, None Gynecology/Gynecologic oncology
Puig & Ross P.A.; Moscarino &
Treu LLP; and Stoll Keenon Ogden
PLLC

Ernest Han, MD, PhD Vergent Biosciences None None Gynecology/Gynecologic oncology

Jordan Holmes, MD, MPH None None None Radiation oncology

Brooke E. Howitt, MD None Cartography Biosciences; and None Pathology


Santa Ana Bio

Jayanthi Lea, MD None Roche Laboratories, Inc. None Gynecology/Gynecologic oncology

Andrea Mariani, MD None None None Gynecology/Gynecologic oncology

David Mutch, MD None None None Gynecology/Gynecologic oncology

Christa Nagel, MD None None None Gynecology/Gynecologic oncology

Larissa Nekhlyudov, MD, MPH None None None Internal medicine; Survivorship

Mirna Podoll, MD None None None Pathology


a
Ritu Salani, MD, MBA Genentech, Inc.; and Merck & Co., Inc. GlaxoSmithKline; ImmunoGen, Inc.; None Gynecology/Gynecologic oncology
Merck & Co., Inc.; Mersana;
Regeneron Pharmactcls; and
Seattle Genetics, Inc.

John Schorge, MD None Avenge Bio None Gynecology/Gynecologic oncology

Jean Siedel, DO, MS None None None Gynecology/Gynecologic oncology

Rachel Sisodia, MD None None None Gynecology/Gynecologic oncology

Pamela Soliman, MD, MPH Incyte Corporation; and Novartis Amgen Inc.; Clovis Oncology; Eisai None Gynecology/Gynecologic oncology
Pharmaceuticals Corporation Inc.; GlaxoSmithKline; Karyopharm
Therapeutics; and Medscape

Stefanie Ueda, MD None None None Gynecology/Gynecologic oncology

Renata Urban, MDa None None None Gynecology/Gynecologic oncology

Stephanie L. Wethington, MD, MSc AstraZeneca Pharmaceuticals LP AstraZeneca Pharmaceuticals LP; None Gynecology/Gynecologic oncology
and GlaxoSmithKline

Emily Wyse, BA None None None Patient advocate

Catheryn Yashar, MD None None None Radiation oncology

Kristine Zanotti, MD None None None Gynecology/Gynecologic oncology

The NCCN Guidelines Staff have no conflicts to disclose.


a
The following individuals have disclosures relating to employment/governing board, patent, equity, or royalty:
Kristin Bradley, MD: UpToDate
Ritu Salani, MD, MBA: Elsevier, UpToDate
Renata Urban, MD: UpToDate

JNCCN.org | Volume 21 Issue 2 | February 2023 209

You might also like