Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Feb 1.
Published in final edited form as: J Natl Compr Canc Netw. 2023 Feb;21(2):217–226. doi: 10.6004/jnccn.2022.7254

Current Evidence-Based Systemic Therapy for Advanced and Recurrent Endometrial Cancer

Sushmita Gordhandas 1,*, William A Zammarrelli III 1,*, Eric V Rios-Doria 1, Angela Green 2,3, Vicky Makker 2,3
PMCID: PMC10361357  NIHMSID: NIHMS1917403  PMID: 36791759

Abstract

Endometrial cancer (EC) is the most common gynecologic malignancy, with worldwide rising incidence and disease-associated mortality. While most patients with EC are diagnosed with early-stage disease, systemic treatment options for patients with advanced or recurrent EC have historically been limited. EC-focused clinical trials and the ensuing therapeutic landscape have expanded since The Cancer Genome Atlas (TCGA) identified four distinct EC subgroups associated with differential survival. This endeavor revolutionized our understanding of the genomic characterization of EC as well as molecular drivers of this heterogeneous malignancy, leading to precision oncology approaches to therapeutics and advancement in treatment options. This manuscript describes the current status of and recent advancements in therapeutic options for patients with advanced and recurrent EC. The National Comprehensive Cancer Network (NCCN) Clinical Practice Guidelines for Uterine Neoplasms Version 1.2022 provides detailed recommendations regarding the diagnosis, work-up, and management of EC.

Introduction

Endometrial cancer (EC) is the most common gynecologic cancer in high-income countries, with the highest incidence in North America. (1) Worldwide, the overall incidence has increased by over 130% in the last 30 years, and disease-associated mortality in the US is rising 2.5% annually on average. (2, 3) The worsening mortality rate is pronounced among Black women, for reasons including decreased healthcare access, treatment disparities, and molecular tumor differences. (4, 5)

Obesity, a strong risk factor for EC, accounts for approximately 50% of cases in Europe and the US. (6) Obesity may contribute to EC pathogenesis through insulin resistance and abnormalities of insulin growth factor 1 signaling, resulting in increased cell proliferation, chronic low-grade inflammation, alterations in adipokine physiology, and cellular and vascular perturbations that promote oncogenesis. (7) Excess endogenous or exogenous estrogen exposure from early menarche, late menopause, or tamoxifen exposure also increase EC risk. Individuals with Lynch syndrome and Cowden syndrome have genetic increased risk of EC, and there is emerging data indicating a potential role for BRCA1/2 germline pathogenic variants in the development of high-grade EC. (8, 9)

Recent studies have highlighted the molecular underpinnings of EC and associated outcomes. (10, 11) The Cancer Genome Atlas (TCGA) Research Network has identified the following four widely accepted molecular EC subtypes: POLE ultra-mutated, mismatch repair (MMR)-deficient (MMRd), copy-number low, and copy-number high (CN-H). (10, 11) Each molecular subtype has its own prognostic significance, with the POLE subtype associated with the best and the CN-H subtype with the poorest survival. (12) While histology and stage are currently used to stratify patients for treatment and to predict outcomes, recent studies have found considerable histologic diversity within EC subtypes, underscoring the importance of the molecular classification of all EC tumors. (12) Molecular subtyping has informed significant therapeutic advancements in advanced/recurrent EC, as outlined in this review, and has been increasingly exploited for risk stratification for optimal personalized treatment. The largest cohort of patients with endometrial cancer who have undergone complete molecular characterization was recently presented by investigators at Memorial Sloan Kettering Cancer Center (Figure 1). Although these patients were treated at a specialized referral center and are disproportionately at higher risk than the general endometrial cancer population, Figure 1 depicts the heterogeneity of patients with endometrial cancer.

Figure 1. Endometrial cancer molecular subtypes and histologies.

Figure 1.

Molecular subtyping of 1906 endometrial cancers at Memorial Sloan Kettering Cancer Center. All four molecular subtypes consist of varying proportions of endometrial cancer histologies. Results presented at the Society for Gynecologic Oncology Annual Meeting, 2022, by Rios-Doria et al., Phoenix, AZ. (12)

These results are preliminary and will be published in finalized form.

CN-H: copy-number high; CN-L: copy-number low; Dediff.: dedifferentiated; Endo.: endometrioid; FIGO: International Federation of Obstetrics and Gynecology; G1: grade 1; G2: grade 2; G3: grade 3; MSI-H: microsatellite instability high; POLE: polymerase epsilon hypermutated; Undiff.: undifferentiated.

While most patients with EC present with early-stage disease and have favorable prognoses, patients with stage III and IV disease have 5-year survival rates of 48% and 15%, respectively. (1) At diagnosis, approximately 20-30% of ECs are considered high risk based on traditional prognostic factors, including stage, histology, myometrial invasion, and lymphovascular invasion, and are associated with recurrence rates of up to 30%. Systemic therapy for advanced/recurrent EC is the cornerstone of treatment. In this review, we will discuss evidence-based therapeutic options for advanced/recurrent EC and review ongoing investigational therapies that may impact the future treatment landscape. Systemic treatment in advanced/recurrent EC remains palliative; therefore, patient quality of life must be prioritized within the shared decision-making process.

First-line (1L) therapy of newly diagnosed advanced EC

The standard 1L therapy for newly diagnosed advanced/recurrent EC regardless of molecular subtype consists of carboplatin/paclitaxel (TC) chemotherapy with or without primary surgical cytoreduction. Key clinical trials assessing systemic therapy for advanced/recurrent EC are summarized in Table 1. TC was established as the standard 1L regimen based on findings from the phase III, randomized non-inferiority Gynecologic Oncology Group (GOG) 209 trial. TC demonstrated non-inferiority over adriamycin, cisplatin, and paclitaxel (TAP) in patients with stage III/IV or recurrent chemotherapy-naïve EC, with an overall survival (OS) of 37 months versus 41 months, respectively (90% CI: 0.9-1.12). TC was also associated with significantly fewer adverse events (AEs) (p<0.05). (13)

Table 1.

Key clinical trials assessing systemic therapy for advanced/recurrent endometrial cancer.

Trial Key Inclusion Criteria Treatment Arms Endpoint(s) Key Findings/Conclusion
First-line
GOG 122
PMID: 16330675

Enrollment: 05/1992-02/2000
-Stage III/IV endometrial carcinoma
-No prior chemotherapy or radiotherapy
-Complete surgical staging
-No single site of residual tumor >2cm
Arm 1: Whole-abdominal irradiation (30 Gy in 20 fractions, with 15-Gy boost); n=202

Arm 2: Doxorubicin + cisplatin n=194
Primary
-PFS
Secondary
-OS
-Arm 2 had improved PFS (HR 0.71; 95% CI 0.55-0.91)
-Arm 2 had improved OS (HR 0.68; 95% CI 0.52-0.89)
-Arm 2 had more grade 3/4 adverse events
GOG 163
PMID: 15277255

Enrollment: 08/12/1996-11/30/1998
-Stage III/IV or recurrent endometrial carcinoma
-No prior cytotoxic chemotherapy
Arm 1: Doxorubicin (60mg/m2) + cisplatin (50mg/m2); n=157

Arm 2: Doxorubicin (50mg/m2) + paclitaxel (150mg/m2) + filgrastim; n=160
Primary
-ORR
Secondary
-PFS
-OS
-No difference in ORR (OR 1.12; 95% CI 0.69-1.79)
-No difference in PFS (HR 1.01; 95% CI 0.8-1.28)
-No difference in OS (HR 1.00; 95% CI 0.78-1.27)
GOG 177
PMID: 15169803

Enrollment: 12/28/1998-08/14/2000
-Stage III/IV or recurrent endometrial carcinoma
-No prior cytotoxic chemotherapy
Arm 1: Doxorubicin (60mg/m2) + cisplatin (50mg/m2); n=129

Arm 2: Doxorubicin (45mg/m2) + cisplatin (50mg/m2) + paclitaxel (160mg/m2) with filgrastim; n=134
Primary
-OS
Secondary
-PFS
-ORR
-Arm 2 had improved PFS (HR 0.60; 95%CI 0.46-0.78)
-Arm 2 had improved OS (HR 0.75; 95% CI 0.57-0.988)
GOG 209
PMID: 33078978

Enrollment: 8/25/2003-4/20/2009
-Stage III/IV or recurrent endometrial carcinoma with poor potential for cure by surgery and/or radiation therapy
-No prior cytotoxic chemotherapy
Arm 1: Paclitaxel (160mg/m2) + doxorubicin (45mg/m2) + cisplatin (50mg/m2); n=640

Arm 2: carboplatin (AUC 6) + paclitaxel (160mg/m2); n=664
Primary
-OS
Secondary
-PFS
-Toxicity
-HRQoL
-Arm 2 was not inferior: OS (HR 1.00; 90%CI 0.9-1.12), PFS (HR 1.03; 90% CI 0.93-1.15)
-Arm 2 had a more favorable toxicity profile and HRQoL difference
PORTEC 3
PMID: 29449189

Enrollment: 11/26/2006-12/20/2013
-Stage IA or IB grade 3 endometrioid endometrial carcinoma, stage II/III endometrioid endometrial carcinoma, or stage IB-III serous or clear cell endometrial cancer
-No prior chemotherapy or radiotherapy
Arm 1: Cisplatin (50mg/m2, 2 cycles) with EBRT followed by carboplatin (AUC 5, 4 cycles); n=330

Arm 2: EBRT; n=330
Primary
-OS
-FFS
Secondary
-Rec rate
-Toxicity
-HRQoL
-No difference in 5-year OS (HR 0.76; 95% CI 0.54-1.06)
-Arm 1 had significantly more Grade 3 complications vs Arm 2 (60% vs 12%, p<0.01)
GOG 258
PMID: 31189035

Enrollment: 06/29/2009-07/28/2014
-Stage III/IVA endometrial carcinoma or stage I/II clear cell/serous endometrial carcinoma with positive washings
-Complete surgical staging, pelvic/paraaortic LN biopsy or dissection was optional
-No single site of residual tumor >2cm
Arm 1: Cisplatin (50mg/m2, 2 cycles) with EBRT followed by carboplatin (AUC 5-6) + paclitaxel (175mg/m2); n=370

Arm 2: Carboplatin (AUC 6) + paclitaxel (175mg/m2); n=360
Primary
-RFS
Secondary
-OS
-Toxicity
-QOL
-No difference in RFS (HR 0.90; 90% CI 0.74-1.10)
-Arm 1 had decreased vaginal recurrences (HR 0.36; 95% CI 0.16-0.82)
-Arm 1 had decreased pelvic/paraaortic LN recurrences (HR 0.43; 95% CI 0.28-0.66)
-Arm 1 had increased distant recurrences (HR 1.36; 95% CI 1.00-1.86)
-Similar incidence of grade 3 or higher toxicity
GOG 261
PMID: 35007153

Enrollment: 8/17/2009-03/24/2014
-Any stage or recurrent uterine or ovarian carcinosarcoma
-Prior radiation therapy allowed but must have been discontinued
Arm 1: Paclitaxel (175mg/m2) + carboplatin (AUC 6); n=224 (uterine)

Arm 2: Ifosfamide (1.6g/m2)+ mesna + paclitaxel (135mg/m2)+ filgrastim; n=204 (uterine)
Primary
-OS
Secondary
-PFS
-AEs
-QOL
-Arm 1 was not inferior to Arm 2
-Arm 1 had improved OS (HR 0.87; 90% CI 0.70-1.08)
-Arm 1 had improved PFS (HR 0.74; 95% CI 0.58-0.93)
-Arm 1 had more hematologic toxicities
-Arm 2 had more confusion and genitourinary hemorrhage
GOG 184
PMID: 19108877

Enrollment: 7/3/2000-9/13/2004
-Stage III/IV endometrial carcinoma
-No prior chemotherapy or radiotherapy
-No single site of residual tumor >2cm
Arm 1: Cisplatin (50mg/m2) + doxorubicin (45mg/m2) + tumor directed radiotherapy; n=288

Arm 2: Cisplatin (50mg/m2) + doxorubicin (45mg/m2) + paclitaxel (160mg/m2) + tumor directed radiotherapy; n=298
Primary
-RFS
-Arm 2 did not have improved RFS (p=0.21, one-tail)
-Arm 2 was associated with increased toxicity
GOG 86P
PMID: 29804638

Enrollment: 9/14/2009-1/9/2012
-Stage III/IV or recurrent endometrial carcinoma
-No prior cytotoxic chemotherapy
Arm 1: paclitaxel (175mg/m2) + carboplatin (AUC 6) + bevacizumab (15mg/kg); n=116

Arm 2: paclitaxel (175mg/m2) + carboplatin (AUC 5) + temsirolimus (25mg); n=115

Arm 3: ixabepilone (30mg/m2) + carboplatin (AUC 6) + bevacizumab (15mg/m2); n=118
Primary
-PFS1
Secondary
-ORR
-OS
-Safety
-PFS was not significantly increased in any arm compared to historical controls2
-An unplanned analysis of PFS stratified by stage/disease status suggested a benefit for Arm 1 compared to historical controls (HR 0.75, 92% CI 0.58-0.95)
-ORR was not significantly increased in any arm compared to historical controls
Subsequent lines
MITO Group End-2 Trial
PMID: 31677820

Enrollment: 04/2012-06/2014
-Stage III/IV or recurrent endometrial carcinoma with measurable disease (RECIST 1.1)
-Excluded carcinosarcoma
-One prior platinum allowed if last treatment > 6 months from study initiation
Arm 1: Carboplatin (AUC 5) + paclitaxel (175mg/m2); n=54

Arm 2: Carboplatin (AUC 5) + paclitaxel (175mg/m2) + bevacizumab (15mg/kg); n=54
Primary
-PFS
Secondary
-OS
-ORR
-No difference in PFS (HR 0.84; 95% CI 0.5-1.3)
-No difference in OS (HR 0.71; 95% CI 0.31-1.36)
-ORR 53% in Arm 1 and 74% in Arm 2
-Arm 2 had increased cardiovascular toxicity and treatment discontinuation
KEYNOTE 775
PMID: 35045221

Enrollment: 06/11/2018-2/3/2020
-Advanced, recurrent, or metastatic
endometrial carcinoma with measurable disease (RECIST 1.1)
-Excluded carcinosarcoma and sarcoma
-At least one prior platinum therapy
-Two lines prior platinum allowed if one line was given in neoadjuvant setting
-No prior PD-1 or VEGF therapies
Arm 1: Lenvatinib (20mg) + pembrolizumab (200mg); n=411

Arm 2: Physician’s choice of doxorubicin (6omg/m2) or paclitaxel (80mg/m2); n=416
Primary
-PFS
-OS
Secondary
-ORR
-QOL
-Arm 1 had improved PFS (HR 0.56; 95% C, 0.47-0.66)
-Arm 1 had improved OS (HR 0.62; 95% CI 0.51-0.75)
-Arm 1 had more grade 3 adverse events vs Arm 2 (88.9% vs 72.7%)

AEs: adverse events; EBRT: External beam pelvic radiotherapy; ECOG: Eastern Cooperative Oncology Group; FFS: Failure free survival; HRQoL: health related quality of life; ORR: objective response rate; OS: overall survival; PFS: progression free survival; PMID: PubMed ID number; QOL: quality of life; RFS: recurrence/relapse free survival; RR: response rate; QOL: quality of life; WHO: World Health Organization.

1

Comparable patients on the PC Arm of trial GOG209 were used as historical controls.

2

Treatment HRs (92% CI) for Arms 1, 2, and 3 relative to controls were 0.81 (0.63–1.02), 1.22 (0.96–1.55) and 0.87 (0.68–1.11), respectively.

In patients with newly diagnosed advanced EC, primary surgical cytoreduction is considered when optimal cytoreduction (to <1 cm residual disease), or preferably, complete gross resection (no visible residual disease) is feasible. However, this approach is largely based on retrospective data, and there is limited high-quality evidence for cytoreduction in advanced EC; recommendations are often extrapolated from ovarian cancer (OVCA) literature. Trends in the upfront treatment of advanced EC are changing, and neoadjuvant chemotherapy is now more commonly used than primary surgical cytoreduction for stage IVB endometrial cancer according to a recent analysis of SEER data. A neoadjuvant chemotherapy approach for advanced EC has been associated with less surgical morbidity and no difference in OS compared to primary surgical cytoreduction. (14, 15)

For locally advanced EC, radiation is routinely used with chemotherapy to reduce the risk of pelvic relapse based on the results of two landmark clinical trials—GOG 258 and PORTEC-3—the latter of which demonstrated a survival benefit with chemoradiation (CTRT) over radiation alone (RT) for stage 3 EC (OS, 78.7% vs. 69.8%, respectively; p=0.114).

Recurrent EC

Chemotherapy

Retrospective data suggest platinum re-challenge has diminished efficacy. Platinum sensitivity, defined as a platinum-free interval of >6 months in OVCA, is terminology that has not been prospectively evaluated or validated in EC. A retrospective analysis of patients retreated with TC who recurred >6 months after completion of adjuvant TC showed an overall response rate (ORR) of 50%, median progression-free survival (PFS) of 10 months, and median OS of 27 months. (16) Retrospective studies have found time to recurrence after prior chemotherapy is predictive of survival in EC. The largest of these studies, a pooled analysis of phase III GOG studies (GOG 107, 122, 163, and 177), which assessed second-line (2L) chemotherapy following primary platinum chemotherapy, showed time to progression after primary platinum chemotherapy was the most predictive factor for survival following 2L chemotherapy. Patients with a platinum-free-interval >6 months compared with <6 months had a 30% lower risk for death (HR, 0.70; 95% CI: 0.59-0.84; p<0.01). (17) There is limited evidence for platinum-based combinations versus single-agent chemotherapy in the 2L or beyond. Following frontline TC, the benefit of subsequent chemotherapy is modest, with a median PFS of 3-4 months and OS of 6-12 months. (1)

The anti–vascular endothelial growth factor (VEGF) antibody bevacizumab is also active in advanced EC as monotherapy and has been explored in combination with TC. The randomized, phase II NRG/GOG 86P trial compared 1L TC plus bevacizumab; TC plus temsirolimus; and ixabepilone and carboplatin plus bevacizumab, and found improved median OS with the addition of bevacizumab to TC compared with historic data from GOG 209 (34 vs 23 months, respectively). Improvements in OS with bevacizumab were most robust among patients with a TP53 mutation on next-generation sequencing or p53 overexpression on immunohistochemistry (IHC), with a median OS for TC plus bevacizumab of 30.0 months compared to 14.4 months for TC plus temsirolimus in this TP53-altered subgroup. (18) The MITO Group End-2 trial looked at TC +/− bevacizumab in patients who had progressed >6 months after completion of 1L therapy. Patients in the bevacizumab arm had a higher ORR, median PFS, and OS, although these differences were not statistically significant. (19) At this time, definitive evidence for the addition of bevacizumab to standard chemotherapy is limited.

Options for single-agent chemotherapy are limited and include agents such as liposomal doxorubicin, doxorubicin, cisplatin, topotecan, docetaxel, and paclitaxel, with ORRs ranging from 9.5-15%. (2025) The National Comprehensive Cancer Network (NCCN) guidelines also include single-agent topotecan, as well as combinations that contain cisplatin, docetaxel, and ifosfamide, all with poor or undocumented efficacy. (21)

Human epidermal growth factor receptor 2 (HER2)

HER2 is emerging as a promising target for the treatment of EC. HER2 is overexpressed in approximately 25-30% of CN-H, serous/serous-like ECs and 13% of carcinosarcomas. When HER2 is overexpressed, tyrosine kinase becomes constitutively activated, leading to dysregulation of DNA transcription through activation of PIK3CA/AKT/mTOR and RAS/RAF/MAPK pathways. Findings from a randomized, phase II trial of TC versus TC plus the monoclonal HER2-targeting antibody trastuzumab in patients with HER2+ stage III/IV or recurrent serous EC demonstrated improved median PFS (12.9 vs 8.0 months, respectively) and OS (29.6 vs 24.4 months, respectively) with TC plus trastuzumab. This benefit was most notable in stage III/IV disease with trastuzumab as part of primary treatment. (26) As a result, TC plus trastuzumab is now a recommended regimen for HER2-overexpressed EC. (27)

Early results of a phase II clinical trial assessing the efficacy and safety of the antibody-drug conjugate (ADC) trastuzumab deruxtecan in HER2-expressing, unresectable, chemotherapy-refractory uterine carcinosarcoma demonstrated a median PFS in IHC HER2 2+/3+ of 6.2 months (95% CI: 4.0-8.8). (28) Trastuzumab deruxtecan is also being investigated in an ongoing phase II study in HER2-expressing endometrial cancer (NCT04482309). Other ADCs currently being investigated in phase I/II studies include HER2-targetting DB-1303 (NCT05150691), folate receptor alpha (FRα)-targeting farletuzumab ecteribulin (NCT04300556), and mirvetuximab soravtansine (IMGN853) monotherapy (NCT03832361) and in combination with pembrolizumab (NCT03835819) (Table 2).

Table 2.

Active/ongoing trials for advanced/recurrent endometrial cancer.

Trial Study start date Recruitment status Treatment arms Experimental maintenance therapy Molecular assessment Estimated enrollment Primary endpoint(s)
First-line Phase 3 Immunotherapy
MK-3475-C93/KEYNOTE-C93/GOG-3064/ENGOT-en15
(NCT05173987)
February 3, 2022 Recruiting - Comparator: Carboplatin + paclitaxel
- Experimental: Pembrolizumab
Pembrolizumab MMRd/MSI N=350
(MMRd only)
PFS, OS
MK-3475-B21/KEYNOTE-B21/ENGOT-en11/GOG-3053
(NCT04634877)
January 10, 2021 Recruiting - Comparator: Carboplatin + paclitaxel + placebo +/− EBRT +/− VBT1
- Experimental: Carboplatin + paclitaxel + pembrolizumab +/− EBRT +/− VBT1
Pembrolizumab MMRd/MSI N=990 DFS, OS
DUO-E
(NCT04269200)
May 21, 2020 Recruiting - Comparator Arm A: Carboplatin + paclitaxel + placebo
- Experimental Arm B: Carboplatin + paclitaxel + durvalumab
- Experimental Arm C: Carboplatin + paclitaxel + durvalumab + olaparib
- Arm B: Durvalumab
- Arm C: Durvalumab + olaparib
MMRd/MSI N=699 PFS
NRG-GY018
(NCT03914612)
July 16, 2019 Recruiting - Comparator: Carboplatin + paclitaxel + placebo
- Experimental: Carboplatin + paclitaxel + pembrolizumab
Pembrolizumab MMRd/MSI N=590 (MMRp)
N=220 (MMRd)
PFS
RUBY
(NCT03981796)
July 18, 2019 Active, not recruiting - Comparator: Carboplatin + paclitaxel + placebo
- Experimental Part 1: Carboplatin + paclitaxel + dostarlimab
- Experimental Part 2: Carboplatin + paclitaxel + dostarlimab + niraparib
- Part 1: Dostarlimab
- Part 2: Dostarlimab + niraparib
MMRd/MSI N=785 PFS, OS
ENGOT-EN9/LEAP-001
(NCT03884101)
April 11, 2019 Active, not recruiting - Comparator: Carboplatin + paclitaxel
- Experimental: Pembrolizumab + lenvatinib
Pembrolizumab MMRd/MSI N=875 PFS, OS
AtTEnd
(NCT03603184)
October 2, 2018 Active, not recruiting - Comparator: Carboplatin + paclitaxel + placebo
- Experimental: Carboplatin + paclitaxel + atezolizumab
Atezolizumab MMRd/MSI N=550 PFS, OS
First-line Phase 3 Non-Immunotherapy
NRG-GY026
(NCT05256225)
August 12, 2022 Recruiting - Comparator Arm I: Carboplatin + paclitaxel
- Experimental Arm II: Carboplatin + paclitaxel + trastuzumab
- Experimental Arm III: Carboplatin + paclitaxel + pertuzumab/trastuzumab
- Arm II: Trastuzumab
- Arm III: Pertuzumab/ trastuzumab
HER2 N=525
(HER2+ serous carcinoma or carcinosarcoma only)
PFS, OS
XPORT-EC
(NCT05611931)
November 1, 2022 Recruiting - Completed a single line of platinum-based therapy
Comparator: Placebo
Experimental: Selinexor
Selinexor TP53 wt N=220 PFS
Phase 2
DESTINY-PanTumor02
(NCT04482309)
June 16, 2023 Active, not recruiting Trastuzumab deruxtecan HER2 N=268 ORR
IMGN853-
(NCT03832361)
July 15, 2020 Recruiting Mirvetuximab soravtansine FRα N=50 ORR
18-602 (NCT03835819) January 2, 2020 Recruiting Mirvetuximab soravtansine + pembrolizumab FRα and MMRp N=35 ORR, PFS
16-322
(NCT02912572)
November 14, 2016 Recruiting - Avelumab
- Avelumab + talazoparib
MSI and/or POLE N=105 ORR, PFS
18-301
(NCT03675893)
December 24, 2018 Recruiting Letrozole + abemaciclib ER-positive N=40 ORR, PFS
INCMGA 0012-204
(NCT04463771)
January 26, 2021 Recruiting - Retifanlimab
- Retifanlimab + epacadostat
- Retifanlimab + pemigatinib
- Retifanlimab + INCAGN02385 + INCAGN2390
MSI, POLE, PD-L1, FGFR1/2/3 N=300 ORR
21-447
(NCT05156268)
January 27, 2022 Recruiting Pembrolizumab + olaparib p53 N=25 ORR
NRG-GY012
(NCT03660826)
September 4, 2018 Recruiting - Arm I: Cediranib
- Arm II: Olaparib
- Arm III: Cediranib + olaparib
- Arm IV: Capivasertib + olaparib
- Arm V: Durvalumab + olaparib
- Arm VI: Cediranib + durvalumab
DNA homologous repair gene mutations N=168 PFS
NRG-GY025
(NCT05112601)
February 7, 2022 Recruiting - Experimental Arm I: Ipilimumab + nivolumab
- Active comparator Arm II: Nivolumab
MMRd/MSI N=12 PFS
NU 18G07
(NCT04049227)
August 12, 2019 Recruiting Abemaciclib + letrozole MMR, PTEN, cyclin D1, p16, pRB N=27 Ki-67 expression
ZN-c3-004
(NCT04814108)
June 1, 2021 Recruiting ZN-c3 N=110 ORR
Phase 1/2
MORAb-202
(NCT04300556)
August 6, 2020 Recruiting Farletuzumab ecteribulin FRα N=55 Dose escalation/ confirmation
DB-1303
(NCT05150691)
January 31, 2022 Recruiting DB-1303 HER2 N=360 Dose escalation/ confirmation
ZN-c3-001
(NCT04158336)
November 1, 2019 Recruiting ZN-c3 N=110 Safety/ tolerability, ORR

C: cycles; DFS: disease-free survival; EBRT: external-beam radiation therapy; ER: estrogen receptor; FRα: folate receptor alpha; HER2: human epidermal growth factor receptor 2; MMRd: mismatch repair–deficient; MMRp: mismatch repair–proficient; MSI: microsatellite instability; NCT: National Clinical Trial identification number; ORR: objective response rate; OS: overall survival; PFS: progression-free survival; TP53 wt: TP53 wildtype; VBT: vaginal brachytherapy.

1

The standard of care radiotherapy regimen may include, at the discretion of the investigator, EBRT ≥4500 cGY with variable dose frequency, with or without cisplatin 50 mg/m2 IV on days 1 and 29 of EBRT, and/or VBT.

MMRd/microsatellite instability–high/tumor mutational burden–high

Approximately 17-33% of advanced and recurrent ECs are MMRd/MSI-high (MSI-H). (29, 30) In MMRd, MSI-H, and tumor mutational burden–high (TMB-H; >10 mutations/megabase) recurrent EC, the recommended 2L systemic therapy is the anti–programmed cell death protein 1 (PD-1) monoclonal antibody pembrolizumab. (31) Other recommended PD-1/programmed death-ligand 1 (PD-L1) agents for the treatment of MMRd/MSI-H EC include dostarlimab, nivolumab, and avelumab. (32, 33)

The Food and Drug Administration (FDA) approval of pembrolizumab for treatment of MMRd/MSI-H EC was largely based on the findings of the phase II KEYNOTE-158 study in patients with advanced MMRd/MSI-H EC, which demonstrated an ORR of 48% and median PFS of 13 months. (34) In 2021, dostarlimab was granted accelerated FDA approval for MMRd/MSI-H EC based on the results of the phase I/II GARNET study, which demonstrated an ORR of 42% and median PFS of 8.1 months in this setting. (32) The PD-1 inhibitor nivolumab has not yet been granted FDA approval but can be used in MMRd/MSI-H EC based on the results of The National Cancer Institute Molecular Analysis for Therapy Choice trial, which showed a 12-month PFS rate of 46.2% in patients with MMRd EC of endometrioid and carcinosarcoma histologies. (33)

MSI is a clear biomarker for response to immune checkpoint inhibitors in EC. However, the underlying mechanisms leading to MSI vary and include germline (Lynch syndrome) and somatic (Lynch-like) MMR gene alterations and sporadic alterations associated with MLH1 methylation. The effect of the underlying mechanism for MSI on immune checkpoint response is unclear. In a phase II study, pembrolizumab was evaluated in recurrent Lynch, Lynch-like, and sporadic MLH1-methylated EC, demonstrating 3-year PFS and OS rates of 100% in Lynch/Lynch-like patients versus 30% and 43%, respectively, in sporadic MLH1-methylated EC. (35)

In the sporadic MLH1-methylated group, mechanism of resistance to PD-L1 agents was associated with acquired loss of beta-2 microglobulin and JAK3, suggesting a lack of surface HLA-class 1 expression and defective response to induction with type I and II interferons. Higher TMB and greater infiltration of CD-68+ macrophages in EC have been proposed as potential mechanisms of response to immunotherapy. (35, 36) While predictive of immune checkpoint response in other cancers, PD-L1 expression is not predictive of response in EC. (37) The anti–PD-L1 agents avelumab and durvalumab have also shown promise as monotherapy in phase II studies in advanced MMRd/MSI-H EC, with ORRs of 27% (and 47%, respectively. (38, 39)

The benefit of incorporating immunotherapy earlier in the therapeutic course, as 1L treatment, is under investigation. There are several ongoing phase III trials of cytotoxic chemotherapy alone or in combination with poly (ADP-ribose) polymerase (PARP) inhibitors, checkpoint inhibitors, and/or VEGF tyrosine kinase inhibitors (TKIs) in 1L therapy of EC, with pending results (Table 2). Whether combination immune checkpoint blockade is more effective than checkpoint inhibitor monotherapy in MMRd/MSI-H EC is of great interest. The phase II NRG-GY025 (NCT05112601) trial of nivolumab versus nivolumab/ipilimumab in recurrent MMRd EC aims to address this question.

MMR-proficient

Most patients with EC have MMR-proficient (MMRp) tumors, which have limited response to single-agent checkpoint inhibitors (ORR, 3-14%). (32, 3739) However, recent studies have established the combination of the multikinase inhibitor lenvatinib plus pembrolizumab as an effective 2L treatment option. Findings from the phase Ib/II Study 111/KEYNOTE 146 trial in patients with advanced EC demonstrated an ORR at week 24 (ORRWk24) of 38.0%. The ORRWk24 was 63.6% for MSI-H tumors (n=11) and 36.2% for microsatellite-stable tumors (n=94). Regardless of MSI status, the median duration of response was 21.2 months, median PFS was 7.4 months, and median OS was 16.7 months in previously treated patients. Responses were seen regardless of PD-L1 expression or histology. (40) These results led to the FDA accelerated approval of lenvatinib plus pembrolizumab for advanced EC that is not MSI-H/MMRd and has progressed following prior systemic therapy.

In the confirmatory phase III Study 309/KEYNOTE775 trial, patients with recurrent EC with measurable disease and one prior platinum-based chemotherapy were enrolled regardless of MMR status. Patients were stratified based on MMR status and randomized 1:1 to lenvatinib plus pembrolizumab or investigator’s choice of doxorubicin or paclitaxel. The median PFS was longer with lenvatinib plus pembrolizumab compared with chemotherapy (MMRp population: 6.6 vs. 3.8 months; p<0.001; overall: 7.2 vs. 3.8 months; p<0.001). The median OS was also longer (MMRp population: 17.4 vs. 12.0 months; p<0.001; overall: 18.3 vs. 11.4 months; p<0.001). PFS and OS analyses in all subtypes, including MMR status, histology, and prior lines of therapy, favored lenvatinib plus pembrolizumab; no substantial differences in health-related quality of life scores were appreciated. (20)

Endocrine therapy

In hormone (estrogen or progesterone)-receptor positive EC, endocrine therapy is a viable treatment option for advanced or recurrent EC. In a translational study of advanced EC tissues from study GOG 119, 40% and 45% of the 45 evaluable tumors were estrogen- and progesterone-receptor positive, respectively. (41) Estrogen and progesterone receptors are typically evaluated with IHC, though timing of when this should be performed is not well established. Current NCCN guidelines recommend hormone receptor testing for stage III, IV, or recurrent endometrioid carcinoma. (21)

Endocrine therapy is generally well tolerated and can be considered in patients with minimal symptoms and low-grade or more indolent disease. A systematic review from 2007 reported that up to 30% of advanced or recurrent ECs respond to endocrine therapy, with the highest response rates in low-grade EC (up to 56% in grade 1/2 endometrioid EC). (42) In the phase II GOG 119 study in advanced EC, alternating tamoxifen and megestrol acetate demonstrated an ORR of 27%, median PFS of 2.7 months, and OS of 14.0 months. (43) Other endocrine therapies include progestational agents alone (medroxyprogesterone acetate or megestrol acetate), aromatase inhibitors, tamoxifen alone, or fulvestrant, a selective estrogen receptor degrader. (21, 44)

Anti-VEGF Therapy

Beyond the combination of bevacizumab with cytotoxic chemotherapy, anti-VEGF therapies have been studied as monotherapy and in combination with other agents in advanced EC. In GOG 229E, bevacizumab monotherapy in previously treated EC was associated with an ORR of 13.5%; and 40.4% of patients were progression-free at 6 months. (45) Lenvatinib monotherapy in a phase II trial in the 2L setting was associated with an ORR of 13.3% and median PFS of 5.6 months. (46)

The NRG-GY012 protocol compared the efficacy of olaparib (PARP inhibitor) monotherapy, cediranib (VEGF receptor TKI) monotherapy, and combination olaparib/cediranib in advanced EC who had received at least one prior line of platinum-based chemotherapy and no more than two prior lines of chemotherapy; median PFS was 2.0, 3.8, and 5.5 months, respectively. (47) Findings from a randomized, phase II trial in recurrent EC demonstrated median PFS for combination nivolumab/cabozantinib (a TKI) and single-agent nivolumab of 5.3, and 1.9 months, respectively. The ORRs were 25% and 16.7%, respectively. (48)

Cell signaling–targeted therapy

Cyclin-dependent kinase inhibitors (CDKis) such as palbociclib and abemaciclib function by selectively inhibiting CDK4 and CDK6, which are crucial for the cell cycle G1/S phase transition. (49) A randomized, phase II trial of letrozole with/without palbociclib (PALEO trial) demonstrated improved PFS with the combination (8.3 vs. 3.0 months; HR, 0.56; p=0.04). (50) Data from a phase II trial in patients with ER+ recurrent EC demonstrated an ORR of 30% and median PFS of 9.1 months with abemaciclib plus letrozole at a median follow-up of 12.5 months. (51) These studies suggest a role for CDK4/6 inhibitors in subsets of advanced ECs, warranting further investigation, which is the subject of ongoing trials (Table 2)

Wee1 Kinase Inhibitors

Wee1 kinase regulates cell-cycle checkpoints of G2/M and S phase. TP53-mutant cancers such as serous EC are often disregulated at the G1/S phase checkpoint, allowing for early S phase entry, rendering them more vulnerable to Wee1 inhibition. Adavosertib is a highly selective inhibitor of Wee1 kinase that in a phase II study in recurrent EC resulted in an ORR of 30%, median PFS of 6.1 months, and median duration of response of 9 months. (52) Additional Wee1 inhibitors are being evaluated in ongoing studies.

Exportin 1 (XPO1) inhibition

Selinexor is an orally available potent inhibitor of XPO1 resulting in retention of tumor suppressor proteins in the nucleus. A phase I open-label study of selinexor with TC in advanced OVCA or EC demonstrated good safety and tolerability. (53 ) A subsequent phase II study of selinexor monotherapy in advanced EC showed a 35% disease control rate. (54) Results of the randomized phase III SIENDO trial of maintenance selinexor vs placebo following response to TC chemotherapy in EC showed the greatest therapeutic benefit in p53 wild-type EC, with a 10-month PFS improvement over placebo. (55) The recently opened phase III XPORT-EC trial (NCT05611931) will evaluate selinexor as maintenance therapy after systemic therapy for p53 wild-type advanced or recurrent EC (Table 2).

Unanswered Questions and Future Directions

Current NCCN guidelines recommend universal testing for MMR proteins in EC to assess for genetic predisposition to Lynch Syndrome and encourage molecular subtyping when available to inform future treatment decisions. (21) While molecular analysis for all ECs is resource intensive, molecular subtyping is considered more predictive of outcomes than other risk stratification criteria in patients with early-stage disease and is anticipated to play an increasing role in adjuvant therapeutic decision making. (5658)

In advanced and recurrent disease, molecular analysis is essential in informing optimal treatment options, delineating predictive biomarkers, and identifying patients eligible for clinical trials investigating emerging therapeutic options based on molecular drivers of EC subtypes. The refinement of MSI-H/MMRd predictive subgroups (e.g., assessment of mutational thresholds and signatures) may identify patients who do not respond or rapidly progress after an initial response to single-agent immune checkpoint inhibition and who could benefit from novel combination strategies. Clinical studies among larger cohorts of Lynch/Lynch-like patients versus sporadic MSI-H patients treated with immune checkpoint inhibitors are warranted. The potential for response to immunotherapy after prior immunotherapy also warrants investigation, as does delineation of mechanisms of response to checkpoint inhibitors in the minority of responding MMRp ECs.

Conclusions

Our understanding of EC biology has been greatly expanded by modern molecular characterization, which has defined molecularly and clinically distinct EC subtypes. Given the heterogeneity of this malignancy, patients with advanced disease should undergo molecular profiling to optimize treatment strategies and inform clinical trial eligibility. The therapeutic ceiling with traditional chemotherapy has been reached, and we must look to relevant biologic targets to individualize treatment. While surgical resection or radiation therapy may be appropriate for localized disease, advanced EC requires systemic therapy. Ongoing phase III studies evaluating immunotherapeutic approaches will likely change the standard of care for 1L management of EC, which will necessitate refinement of 2L and beyond management. While the incidence and number of deaths from EC has increased worldwide, we must advocate for research to improve the quality of life and outcomes for patients with EC.

Funding:

Drs. Makker and Green are supported in part through the NIH/NCI Cancer Center Support Grant P30 1CA008748.

Conflict of interest statement

Dr. Makker reports research (all funding to institution)/consultant/advisory board member support from Merck, Eisai, Karyopharm, AstraZeneca, Clovis, Moreo, Takeda, Zymeworks, and Genentech, GSK, Dicephera, Faeth. Dr. Green reports research funding paid to the institution from Mereo Biopharma and Eli Lilly. The other authors have no conflicts of interest to disclose.

References

  • 1.Crosbie EJ, Kitson SJ, McAlpine JN, Mukhopadhyay A, Powell ME, Singh N. Endometrial cancer. The Lancet. 2022;399(10333):1412–28. [DOI] [PubMed] [Google Scholar]
  • 2.Gu B, Shang X, Yan M, Li X, Wang W, Wang Q, et al. Variations in incidence and mortality rates of endometrial cancer at the global, regional, and national levels, 1990–2019. Gynecologic Oncology. 2021;161(2):573–80. [DOI] [PubMed] [Google Scholar]
  • 3.Matsuo K, Mandelbaum RS, Matsuzaki S, Klar M, Roman LD, Wright JD. Ovarian conservation for young women with early-stage, low-grade endometrial cancer: a 2-step schema. American journal of obstetrics and gynecology. 2021;224(6):574–84. [DOI] [PubMed] [Google Scholar]
  • 4.Clarke MA, Devesa SS, Harvey SV, Wentzensen N. Hysterectomy-Corrected Uterine Corpus Cancer Incidence Trends and Differences in Relative Survival Reveal Racial Disparities and Rising Rates of Nonendometrioid Cancers. J Clin Oncol. 2019;37(22):1895–908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Giaquinto AN, Broaddus RR, Jemal A, Siegel RL. The Changing Landscape of Gynecologic Cancer Mortality in the United States. Obstet Gynecol. 2022;139(3):440–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Setiawan VW, Yang HP, Pike MC, McCann SE, Yu H, Xiang YB, et al. Type I and II endometrial cancers: have they different risk factors? J Clin Oncol. 2013;31(20):2607–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Onstad MA, Schmandt RE, Lu KH. Addressing the Role of Obesity in Endometrial Cancer Risk, Prevention, and Treatment. J Clin Oncol. 2016;34(35):4225–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Smith ES, Da Cruz Paula A, Cadoo KA, Abu-Rustum NR, Pei X, Brown DN, et al. Endometrial Cancers in BRCA1 or BRCA2 Germline Mutation Carriers: Assessment of Homologous Recombination DNA Repair Defects. JCO Precision Oncology. 2019(3):1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Cadoo KA, Mandelker DL, Mukherjee S, Stewart C, Delair D, Ravichandran V, et al. Understanding Inherited Risk in Unselected Newly Diagnosed Patients With Endometrial Cancer. JCO Precision Oncology. 2019(3):1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Levine DA. Integrated genomic characterization of endometrial carcinoma. Nature. 2013;497(7447):67–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Cancer Genome Atlas Research N, Kandoth C, Schultz N, Cherniack AD, Akbani R, Liu Y, et al. Integrated genomic characterization of endometrial carcinoma. Nature. 2013;497(7447):67–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Rios-Doria E Molecular Classification of Endometrial Carcinomas: A Single Institution Review. 2022 SGO Annual Meeting on Women’s Cancer; March 18–21, 2022; Phoenix, AZ2022. [Google Scholar]
  • 13.Miller DS, Filiaci VL, Mannel RS, Cohn DE, Matsumoto T, Tewari KS, et al. Carboplatin and Paclitaxel for Advanced Endometrial Cancer: Final Overall Survival and Adverse Event Analysis of a Phase III Trial (NRG Oncology/GOG0209). Journal of Clinical Oncology. 2020;38(33):3841–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Jooya ND, Ciccone MA, Brunette LL, Pham HQ, Yessaian AA, Muderspach LI, et al. Population-level uptake of neoadjuvant chemotherapy for stage IVB endometrial cancer. Gynecol Oncol. 2022;165(3):428–36. [DOI] [PubMed] [Google Scholar]
  • 15.Huang AB, Wu J, Chen L, Albright BB, Previs RA, Moss HA, et al. Neoadjuvant chemotherapy for advanced stage endometrial cancer: A systematic review. Gynecol Oncol Rep. 2021;38:100887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Rubinstein M, Halpenny D, Makker V, Grisham RN, Aghajanian C, Cadoo K. Retreatment with carboplatin and paclitaxel for recurrent endometrial cancer: A retrospective study of the Memorial Sloan Kettering Cancer Center experience. Gynecol Oncol Rep. 2019;28:120–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Moore KN, Tian C, McMeekin DS, Thigpen JT, Randall ME, Gallion HH. Does the progression-free interval after primary chemotherapy predict survival after salvage chemotherapy in advanced and recurrent endometrial cancer?: a Gynecologic Oncology Group ancillary data analysis. Cancer. 2010;116(23):5407–14. [DOI] [PubMed] [Google Scholar]
  • 18.Aghajanian C, Filiaci V, Dizon DS, Carlson JW, Powell MA, Secord AA, et al. A phase II study of frontline paclitaxel/carboplatin/bevacizumab, paclitaxel/carboplatin/temsirolimus, or ixabepilone/carboplatin/bevacizumab in advanced/recurrent endometrial cancer. Gynecol Oncol. 2018;150(2):274–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Lorusso D, Ferrandina G, Colombo N, Pignata S, Pietragalla A, Sonetto C, et al. Carboplatin-paclitaxel compared to Carboplatin-Paclitaxel-Bevacizumab in advanced or recurrent endometrial cancer: MITO END-2 - A randomized phase II trial. Gynecol Oncol. 2019;155(3):406–12. [DOI] [PubMed] [Google Scholar]
  • 20.Makker V, Colombo N, Casado Herráez A, Santin AD, Colomba E, Miller DS, et al. Lenvatinib plus Pembrolizumab for Advanced Endometrial Cancer. New England Journal of Medicine. 2022;386(5):437–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Network NCC. Uterine Neoplasms (Version 1.2022) [Available from: https://www.nccn.org/professionals/physician_gls/pdf/uterine.pdf. [Google Scholar]
  • 22.Traina TA, Sabbatini P, Aghajanian C, Dupont J. Weekly topotecan for recurrent endometrial cancer: a case series and review of the literature. Gynecol Oncol. 2004;95(1):235–41. [DOI] [PubMed] [Google Scholar]
  • 23.Thigpen JT, Buchsbaum HJ, Mangan C, Blessing JA. Phase II trial of adriamycin in the treatment of advanced or recurrent endometrial carcinoma: a Gynecologic Oncology Group study. Cancer Treat Rep. 1979;63(1):21–7. [PubMed] [Google Scholar]
  • 24.Thigpen JT, Blessing JA, Beecham J, Homesley H, Yordan E. Phase II trial of cisplatin as first-line chemotherapy in patients with advanced or recurrent uterine sarcomas: a Gynecologic Oncology Group study. J Clin Oncol. 1991;9(11):1962–6. [DOI] [PubMed] [Google Scholar]
  • 25.Sutton GP, Blessing JA, DeMars LR, Moore D, Burke TW, Grendys EC. A phase II Gynecologic Oncology Group trial of ifosfamide and mesna in advanced or recurrent adenocarcinoma of the endometrium. Gynecol Oncol. 1996;63(1):25–7. [DOI] [PubMed] [Google Scholar]
  • 26.Fader AN, Roque DM, Siegel E, Buza N, Hui P, Abdelghany O, et al. Randomized Phase II Trial of Carboplatin-Paclitaxel Compared with Carboplatin-Paclitaxel-Trastuzumab in Advanced (Stage III-IV) or Recurrent Uterine Serous Carcinomas that Overexpress Her2/Neu (NCT01367002): Updated Overall Survival Analysis. Clin Cancer Res. 2020;26(15):3928–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Tymon-Rosario J, Siegel ER, Bellone S, Harold J, Adjei N, Zeybek B, et al. Trastuzumab tolerability in the treatment of advanced (stage III-IV) or recurrent uterine serous carcinomas that overexpress HER2/neu. Gynecol Oncol. 2021;163(1):93–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Hasegawa K, Nishikawa T, Hirakawa A, Kawasaki M, Tomatsuri S, Nagasaka Y, et al. 813P Efficacy and safety of trastuzumab deruxtecan in HER2-expressing uterine carcinosarcoma (STATICE trial, NCCH1615): A multicenter, phase II clinical trial. Annals of Oncology. 2021;32:S767. [Google Scholar]
  • 29.Marabelle A, Le DT, Ascierto PA, Di Giacomo AM, De Jesus-Acosta A, Delord JP, et al. Efficacy of Pembrolizumab in Patients With Noncolorectal High Microsatellite Instability/Mismatch Repair-Deficient Cancer: Results From the Phase II KEYNOTE-158 Study. J Clin Oncol. 2020;38(1):1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Cortes-Ciriano I, Lee S, Park WY, Kim TM, Park PJ. A molecular portrait of microsatellite instability across multiple cancers. Nat Commun. 2017;8:15180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Kahn RM, Gordhandas S, Maddy BP, Baltich Nelson B, Askin G, Christos PJ, et al. Universal endometrial cancer tumor typing: How much has immunohistochemistry, microsatellite instability, and MLH1 methylation improved the diagnosis of Lynch syndrome across the population? Cancer. 2019;125(18):3172–83. [DOI] [PubMed] [Google Scholar]
  • 32.Oaknin A, Tinker AV, Gilbert L, Samouëlian V, Mathews C, Brown J, et al. Clinical Activity and Safety of the Anti-Programmed Death 1 Monoclonal Antibody Dostarlimab for Patients With Recurrent or Advanced Mismatch Repair-Deficient Endometrial Cancer: A Nonrandomized Phase 1 Clinical Trial. JAMA Oncol. 2020;6(11):1766–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Azad NS, Gray RJ, Overman MJ, Schoenfeld JD, Mitchell EP, Zwiebel JA, et al. Nivolumab Is Effective in Mismatch Repair-Deficient Noncolorectal Cancers: Results From Arm Z1D-A Subprotocol of the NCI-MATCH (EAY131) Study. J Clin Oncol. 2020;38(3):214–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.O’Malley DM, Bariani GM, Cassier PA, Marabelle A, Hansen AR, De Jesus Acosta A, et al. Pembrolizumab in Patients With Microsatellite Instability-High Advanced Endometrial Cancer: Results From the KEYNOTE-158 Study. J Clin Oncol. 2022;40(7):752–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Bellone S, Roque DM, Siegel ER, Buza N, Hui P, Bonazzoli E, et al. A phase 2 evaluation of pembrolizumab for recurrent Lynch-like versus sporadic endometrial cancers with microsatellite instability. Cancer. 2022;128(6):1206–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Bellone S, Roque DM, Siegel ER, Buza N, Hui P, Bonazzoli E, et al. A phase II evaluation of pembrolizumab in recurrent microsatellite instability-high (MSI-H) endometrial cancer patients with Lynch-like versus MLH−1 methylated characteristics (NCT02899793). Annals of Oncology. 2021;32(8):1045–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Ott PA, Bang YJ, Berton-Rigaud D, Elez E, Pishvaian MJ, Rugo HS, et al. Safety and Antitumor Activity of Pembrolizumab in Advanced Programmed Death Ligand 1-Positive Endometrial Cancer: Results From the KEYNOTE-028 Study. J Clin Oncol. 2017;35(22):2535–41. [DOI] [PubMed] [Google Scholar]
  • 38.Konstantinopoulos PA, Luo W, Liu JF, Gulhan DC, Krasner C, Ishizuka JJ, et al. Phase II Study of Avelumab in Patients With Mismatch Repair Deficient and Mismatch Repair Proficient Recurrent/Persistent Endometrial Cancer. J Clin Oncol. 2019;37(30):2786–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Antill Y, Kok PS, Robledo K, Yip S, Cummins M, Smith D, et al. Clinical activity of durvalumab for patients with advanced mismatch repair-deficient and repair-proficient endometrial cancer. A nonrandomized phase 2 clinical trial. J Immunother Cancer. 2021;9(6). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Makker V, Taylor MH, Aghajanian C, Oaknin A, Mier J, Cohn AL, et al. Lenvatinib Plus Pembrolizumab in Patients With Advanced Endometrial Cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2020;38(26):2981–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Singh M, Zaino RJ, Filiaci VJ, Leslie KK. Relationship of estrogen and progesterone receptors to clinical outcome in metastatic endometrial carcinoma: a Gynecologic Oncology Group Study. Gynecol Oncol. 2007;106(2):325–33. [DOI] [PubMed] [Google Scholar]
  • 42.Decruze SB, Green JA. Hormone therapy in advanced and recurrent endometrial cancer: a systematic review. Int J Gynecol Cancer. 2007;17(5):964–78. [DOI] [PubMed] [Google Scholar]
  • 43.Fiorica JV, Brunetto VL, Hanjani P, Lentz SS, Mannel R, Andersen W. Phase II trial of alternating courses of megestrol acetate and tamoxifen in advanced endometrial carcinoma: a Gynecologic Oncology Group study. Gynecol Oncol. 2004;92(1):10–4. [DOI] [PubMed] [Google Scholar]
  • 44.Altman AD, Thompson J, Nelson G, Chu P, Nation J, Ghatage P. Use of aromatase inhibitors as first- and second-line medical therapy in patients with endometrial adenocarcinoma: a retrospective study. J Obstet Gynaecol Can. 2012;34(7):664–72. [DOI] [PubMed] [Google Scholar]
  • 45.Aghajanian C, Sill MW, Darcy KM, Greer B, McMeekin DS, Rose PG, et al. Phase II trial of bevacizumab in recurrent or persistent endometrial cancer: a Gynecologic Oncology Group study. J Clin Oncol. 2011;29(16):2259–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Vergote I, Powell MA, Teneriello MG, Miller DS, Garcia AA, Mikheeva ON, et al. Second-line lenvatinib in patients with recurrent endometrial cancer. Gynecologic Oncology. 2020;156(3):575–82. [DOI] [PubMed] [Google Scholar]
  • 47.Rimel BJ. A randomized, phase ii study comparing single-agent olaparib, single agent cediranib, and the combination of cediranib/olaparib in women with recurrent, persistent or metastatic endometrial cancer. Gynecologic Oncology. 2021;162:S43–S4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Lheureux S, Matei D, Konstantinopoulos PA, Block MS, Jewell A, Gaillard S, et al. A randomized phase II study of cabozantinib and nivolumab versus nivolumab in recurrent endometrial cancer. American Society of Clinical Oncology; 2020. [Google Scholar]
  • 49.Giannone G, Tuninetti V, Ghisoni E, Genta S, Scotto G, Mittica G, et al. Role of Cyclin-Dependent Kinase Inhibitors in Endometrial Cancer. Int J Mol Sci. 2019;20(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Mirza MR, Monk BJ, Herrstedt J, Oza AM, Mahner S, Redondo A, et al. Niraparib Maintenance Therapy in Platinum-Sensitive, Recurrent Ovarian Cancer. N Engl J Med. 2016;375(22):2154–64. [DOI] [PubMed] [Google Scholar]
  • 51.Konstantinopoulos PA. Phase 2, two-stage study of letrozole and abemaciclib in estrogen receptor (ER) positive recurrent or metastatic endometrial cancer (EC). . 2022 SGO Annual Meeting on Women’s Cancer; March 18–21, 2022; Phoenix, AZ2022. [Google Scholar]
  • 52.Liu JF, Xiong N, Campos SM, Wright AA, Krasner C, Schumer S, et al. Phase II Study of the WEE1 Inhibitor Adavosertib in Recurrent Uterine Serous Carcinoma. J Clin Oncol. 2021;39(14):1531–9. [DOI] [PubMed] [Google Scholar]
  • 53.Rubinstein MM, Grisham RN, Cadoo K, Kyi C, Tew WP, Friedman CF, et al. A phase I open-label study of selinexor with paclitaxel and carboplatin in patients with advanced ovarian or endometrial cancers. Gynecol Oncol. 2021;160(1):71–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Vergote IB, Lund B, Peen U, Umajuridze Z, Mau-Sorensen M, Kranich A, et al. Phase 2 study of the Exportin 1 inhibitor selinexor in patients with recurrent gynecological malignancies. Gynecol Oncol. 2020;156(2):308–14. [DOI] [PubMed] [Google Scholar]
  • 55.Makker V, Perez-Fidalgo JA, Bergamini A, Spitz DL, Gorp TV, Sehouli J, et al. Randomized phase III study of maintenance selinexor versus placebo in endometrial cancer (ENGOT-EN5/GOG-3055/SIENDO): Impact of subgroup analysis and molecular classification. Journal of Clinical Oncology. 2022;40(16_suppl):5511-. [Google Scholar]
  • 56.Zammarrelli WA 3rd, Kim SH, Da Cruz Paula A,Rios-Doria EV, Ehmann S, Yeoshoua E, et al. Risk Stratification of Stage I Grade 3 Endometrioid Endometrial Carcinoma in the Era of Molecular Classification. JCO Precis Oncol. 2022;6:e2200194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.van den Heerik A, Horeweg N, Nout RA, Lutgens L, van der Steen-Banasik EM, Westerveld GH, et al. PORTEC-4a: international randomized trial of molecular profile-based adjuvant treatment for women with high-intermediate risk endometrial cancer. Int J Gynecol Cancer. 2020;30(12):2002–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Murali R, Delair DF, Bean SM, Abu-Rustum NR, Soslow RA. Evolving Roles of Histologic Evaluation and Molecular/Genomic Profiling in the Management of Endometrial Cancer. J Natl Compr Canc Netw. 2018;16(2):201–9. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES