ArticlePDF AvailableLiterature Review

Abstract and Figures

The treatment of recurrent endometrial cancer is a challenge. Because of earlier treatments and the site of locoregional recurrence, in the vaginal vault or pelvis, morbidity can be high. A total of about 4 to 20% of the patients with endometrial cancer develop a locoregional recurrence, mostly among patients with locally advanced disease. The treatment options are dependent on previous treatments and the site of recurrence. Local and locoregional recurrences can be treated curatively with surgery or (chemo)radiotherapy with acceptable toxicity and control rates. Distant recurrences can be treated with palliative systemic therapy, i.e., first-line chemotherapy or hormonal therapy. Based on the tumor characteristics and molecular profile, there can be a role for immunotherapy. The evidence on targeted therapy is limited, with no approved treatment in the current guidelines. In selected cases, there might be an indication for local treatment in oligometastatic disease. Because of the novel techniques in radiotherapy, disease control can often be achieved at limited toxicity. Further studies are warranted to analyze the survival outcome and toxicity of newer treatment strategies. Patient selection is very important in deciding which treatment is of most benefit, and better prediction models based on the patient- and tumor characteristics are necessary.
Content may be subject to copyright.
cancers
Review
Recurrent Endometrial Cancer: Local and Systemic
Treatment Options
Heidi Rütten 1, *, Cornelia Verhoef 1, Willem Jan van Weelden 2, Anke Smits 2, Joëlle Dhanis 3,
Nelleke Ottevanger 4and Johanna M. A. Pijnenborg 2


Citation: Rütten, H.; Verhoef, C.; van
Weelden, W.J.; Smits, A.; Dhanis, J.;
Ottevanger, N.; Pijnenborg, J.M.A.
Recurrent Endometrial Cancer: Local
and Systemic Treatment Options.
Cancers 2021,13, 6275. https://
doi.org/10.3390/cancers13246275
Academic Editors: Miguel Abal,
Laura Muinelo-Romay and
Dionyssios Katsaròs
Received: 30 September 2021
Accepted: 8 December 2021
Published: 14 December 2021
Publisher’s Note: MDPI stays neutral
with regard to jurisdictional claims in
published maps and institutional affil-
iations.
Copyright: © 2021 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
1Department of Radiation Oncology, Radboudumc, 6525 GA Nijmegen, The Netherlands;
Lia.Verhoef@radboudumc.nl
2Department of Obstetrics & Gynaecology, Radboudumc, 6525 GA Nijmegen, The Netherlands;
willemjan.vanweelden@radboudumc.nl (W.J.v.W.); Anke.Smits@radboudumc.nl (A.S.);
Hanny.MA.Pijnenborg@radboudumc.nl (J.M.A.P.)
3Faculty of Medical Sciences, Radboud University, Houtlaan 4, 6525 XZ Nijmegen, The Netherlands;
j.dhanis@ru.student.nl
4Department of Medical Oncology, Radboudumc, 6525 GA Nijmegen, The Netherlands;
Nelleke.Ottevanger@radboudumc.nl
*Correspondence: heidi.rutten@radboudumc.nl
Simple Summary:
In this review, we discuss the different treatment strategies in recurrent endome-
trial cancer. The incidence of endometrial cancer is rising. The available treatment options increase
with the development of novel radiotherapy techniques and new systemic therapies. Dependent
on the site of recurrence and previous therapy, the treatment of recurrent endometrial cancer can
be curative or palliative. Newly emerging medical treatments, such as immunotherapy, might be
of benefit in selected patients. Moreover, combinations of different treatments can lead to a better
outcome. Recent insights on oligometastatic disease lead us to expect that ablative or radical local
treatment for distant metastasis will be of benefit in selected patients. Due to the complexity of the
cases, it is recommended to discuss individual cases in a multidisciplinary tumor board. Shared
decision-making principles are recommended to maximize treatment personalization.
Abstract:
The treatment of recurrent endometrial cancer is a challenge. Because of earlier treatments
and the site of locoregional recurrence, in the vaginal vault or pelvis, morbidity can be high. A total
of about 4 to 20% of the patients with endometrial cancer develop a locoregional recurrence, mostly
among patients with locally advanced disease. The treatment options are dependent on previous
treatments and the site of recurrence. Local and locoregional recurrences can be treated curatively
with surgery or (chemo)radiotherapy with acceptable toxicity and control rates. Distant recurrences
can be treated with palliative systemic therapy, i.e., first-line chemotherapy or hormonal therapy.
Based on the tumor characteristics and molecular profile, there can be a role for immunotherapy. The
evidence on targeted therapy is limited, with no approved treatment in the current guidelines. In
selected cases, there might be an indication for local treatment in oligometastatic disease. Because
of the novel techniques in radiotherapy, disease control can often be achieved at limited toxicity.
Further studies are warranted to analyze the survival outcome and toxicity of newer treatment
strategies. Patient selection is very important in deciding which treatment is of most benefit, and
better prediction models based on the patient- and tumor characteristics are necessary.
Keywords:
endometrial cancer; recurrence; treatment; surgery; radiotherapy; hormonal therapy;
systemic treatment; oligometastases
1. Introduction
Endometrial cancer (EC) is the most common gynecological cancer in the Western
world. Its incidence is rising as risk factors for endometrial cancer are more and more
Cancers 2021,13, 6275. https://doi.org/10.3390/cancers13246275 https://www.mdpi.com/journal/cancers
Cancers 2021,13, 6275 2 of 14
prevalent [
1
,
2
]. The risk factors for developing endometrial cancer include prolonged
unopposed estrogen exposure, advanced age, and obesity [
3
]. Most patients are diagnosed
in their sixth or seventh decade, and, since postmenopausal blood loss is one of the first
symptoms, most patients present with early-stage disease [4].
Endometrial cancer can be classified in different ways. Historically, two subtypes
were recognized [
5
]. Type one tumors are the most common (70% of all tumors) and are
predominantly well to moderately differentiated endometroid tumors with often high
expression of the estrogen–progesterone receptor (ER/PR), carrying a good prognosis
with surgery alone. Type two tumors are poorly differentiated endometroid carcinomas
or more aggressive subtypes, such as clear cell or serous carcinomas. Type two tumors
tend to be more advanced at diagnosis and have a poorer prognosis [
6
]. Nowadays, the
classification according to the molecular profile is upcoming with tumor types such as
POLE, MSI, and P53, where the first two predominantly have a favorable prognosis and
the latter is considered to be an aggressive subtype [7,8].
The relationship between the different histological subtypes and molecular profiles is
shown in Figure 1. As illustrated, there is quite some overlap in the historically type two
tumors and the more aggressive molecular subtypes, whereas the type one tumors tend to
have a more favorable molecular profile.
Figure 1.
Relationship between the traditional histologic classification and the molecular classification.
Each traditional histologic diagnosis is connected to the representing molecular subgroup. The
thicker the connecting line, the stronger the relationship. The figure demonstrates that each molecular
subgroup can be detected in each histologic subgroup. Yet, the NSMP is mainly reflected by grade 1
and 2 EEC (left in the figure), whereas the p53abn cancers are mainly reflected by patients with
SC (right in the figure). EEC: endometrioid endometrial cancer; CCC: clear cell carcinoma; SC:
serous cancer; NSMP: nonspecific molecular profile; MMRd: mismatch repair deficient; POLE: POLE
ultramutated; p53abn: copy number high/TP53 mutated. (Modified from [
9
] UpToDate Endometrial
cancer: Pathology and classification by Huvila J, MD, PhD, McAlpine JN, MD, FACOG, FRCPSC,
available from: URL: https://www.uptodate.com/contents/endometrial-cancer-pathology-and-
classification?source=history_widget) accessed 17 September 2021.
The primary treatment consists of the surgical removal of the uterus and adnexa with
or without lymph node dissection, or the sentinel node procedure. The rationale for nodal
staging is based on the risk factors, i.e., tumor grade, deep myometrial invasion, or, with
increasing evidence, molecular profile, but remains a subject of discussion [10,11].
The adjuvant treatment is based on the risk of locoregional recurrence or metastasis
and can be locoregional radiotherapy, chemotherapy, or a combination of both. Patients
Cancers 2021,13, 6275 3 of 14
with low-risk tumors can be treated with surgery alone, whereas patients with high-risk
tumors are eligible for adjuvant chemotherapy and/or radiotherapy. There is no real
international consensus about treating intermediate-risk tumors. The treatment strategies
can vary between surgery only, adjuvant brachytherapy or pelvic radiotherapy, and/or
chemotherapy [10,11].
Despite optimal surgical and adjuvant treatment, 7–15% of early stage (I-II) patients
present with recurrent disease [
12
14
]. This can be locoregional recurrence, distant metas-
tasis, or both. The risk of locoregional recurrence is low [
12
,
15
,
16
] and strongly related to
the presence of risk factors, such as LVSI, tumor grade, or molecular profile [
14
]. About
50% of the patients with a recurrence have locoregional disease, 25% present with distant
recurrence, and the remaining 25% have both [
14
]. Patients with advanced-stage disease at
diagnosis or with a more aggressive subtype have a higher probability of both locoregional
and distant recurrence [
17
]. Several studies showed a relapse-free survival of 60 to 70% after
complete debulking and adjuvant (chemo)radiation in locally advanced disease [
17
,
18
].
Most recurrences occur within three years after the primary treatment, with a median
5-year survival of 55% after pelvic recurrence and 17% after metastatic disease [12,14].
The treatment options are dependent on the site of recurrence, tumor histology, i.e.,
biomarkers, previous treatments, and the patient’s performance status and preferences.
The treatment strategies can include surgery, radiotherapy, hormonal treatment, systemic
treatment, or a combination [
19
]. In this review, we will give an overview of the local and
systemic treatment options for patients with recurrent endometrial cancer and intend to
provide insight regarding the different treatment strategies for patients with an increasingly
personalized approach in the future.
2. Surgical Treatment for Locoregional Recurrence
2.1. Vaginal Vault Recurrence
For a vaginal recurrence in previously irradiated patients, surgical resection is con-
sidered to be the first-line treatment option, followed by adjuvant local radiotherapy (i.e.,
image-guided brachytherapy) if indicated [13,20,21].
Vaginal vault recurrences in non-irradiated patients are traditionally salvaged with
radiation therapy [
13
]. However, the tumor size significantly influences the effect of
radiotherapy and, therefore, surgical resection may be considered as a viable alternative.
Wylie et al. showed that local control with radiotherapy was significantly worse for tumors
>2 cm compared to smaller tumors (54% versus 80%) [
22
]. With the use of a combination
of external beam radiotherapy (EBRT) and brachytherapy, also in larger tumors, a good
locoregional control can be achieved [
23
]. Although Haldarson et al. showed comparable
survival in patients with surgical resection and radiotherapy, in previously non-irradiated
patients, the limited number of patients warrants further research [24].
2.2. Locoregional and Abdominal Recurrence
Historically, the role of surgery for recurrent endometrial cancer has been focused on
exenterative surgery. Pelvic exenteration (anterior, posterior, or total) is performed with
curative intent and has primarily been described in highly selected patients with an isolated
pelvic adenocarcinoma recurrence after radiotherapy [
25
]. The complete resection of the
tumor is reported to be feasible in the majority of patients, with the reported complete
resection rates varying from 86% to 100% in a small case series [
25
27
]. However, the selec-
tion criteria are lacking, and the results are based on a small number of patients that have
been selected retrospectively over long time periods. In addition, resection is accompanied
by significant surgical-related morbidity and mortality with major complications in up to
80% of patients and mortality rates of 5% [
25
,
27
]. The five-year survival rates after pelvic
exenteration are reported to be up to 56 to 70% when complete resection has been obtained
(R0) compared to 20% or less in the presence of residual disease (R1-2) [2527].
Over the past years, the role of surgical cytoreduction (i.e., excision of all visible
disease) for recurrent endometrial cancer has been gaining increasing interest as a surgical
Cancers 2021,13, 6275 4 of 14
alternative with less morbidity compared to exenteration. Retrospective studies have
demonstrated the feasibility and additional value of cytoreductive surgery [
28
32
]. Patient
selection was based upon a local multi-disciplinary team review and included endometroid
and non-endometroid tumors. Most studies included both locoregional (pelvic and nodal)
and intra-abdominal recurrences. After cytoreduction, 40–60% of the patients were treated
with systemic therapy, radiotherapy, or both [
21
,
33
,
34
]. The removal of all visible disease
was achieved in the majority of the patients, varying from 56% to 71% [
21
,
29
,
33
]. The factors
associated with complete cytoreduction were: solitary recurrence, tumor size (<6 cm), and
performance status [
30
,
32
,
33
]. Advanced age and the presence of peritonitis carcinomatosis
negatively impacted the achievement of complete cytoreduction and survival [
32
,
34
]. The
surgical morbidity rates varied from 9% to 21%, mainly grade one and two complications,
and no perioperative deaths were reported [2830,32].
Complete resection was significantly associated with improved overall survival [
21
,
28
,
31
,
32
].
A recent multi-institutional study of 230 patients reported a significantly improved 5-year
survival of 66% in patients with no residual disease and negative resection margins com-
pared to 37% in patients with macroscopic residual disease. The site of recurrence did
not impact the survival outcomes [
31
]. In a large meta-analysis by Barlin et al., the role
of cytoreductive surgery for both advanced (n = 515) and recurrent (n = 157) endometrial
cancer patients was evaluated. Complete cytoreduction was associated with a superior
overall survival outcome, with each 10% increase in proportion with the patients under-
going complete cytoreduction showing a 9-month increase in survival. In the cases with
residual disease of 2 cm or more, the survival benefit was lost, supporting proper selection
and the aim to achieve complete debulking [35].
Whether neoadjuvant chemotherapy in a recurrent setting might be a valuable treat-
ment option prior to debulking has not been studied so far. Based on the data in the
primary setting, neoadjuvant chemotherapy in advanced-stage disease resulted in 80%
complete debulking [
36
] and, as such, could be considered in individual patients with a
good performance status.
2.3. Solitary Distant Metastasis
There are few reports on the surgical management of isolated distant recurrences.
Tangjitgamol et al. reviewed the role of the surgical resection of solitary pulmonary, hepatic,
and cerebral metastasis and deemed it feasible for individualized cases. The successful
resection of splenic metastasis has also been reported [
37
]. The favorable prognostic factors
for a prolonged survival were good performance status, long disease-free interval, and
clear margins [
38
]. However, the studies are limited and comprise mainly case reports,
warranting further research before evidence-based guidelines can be drafted.
3. Radiotherapy
3.1. Vaginal Vault Recurrence
In the PORTEC-1 trial, the locoregional recurrence was 14% in previously unirradiated
patients with 11% vaginal recurrence and 3% pelvic recurrence. In previously irradiated
patients, 4% locoregional relapse occurred, of which 2% was pelvic recurrence [
15
]. This
is in line with other published recurrence rates, such as Francis et al., who found 7%
overall recurrences with 4% only locoregional recurrences in 2691 patients with stage I-II
endometrial cancer [16].
The treatment of a vaginal recurrence usually requires a combination of external
beam radiotherapy (EBRT) with elective nodal irradiation and brachytherapy boost. In
radiation-naïve patients, this is often the treatment of choice. Local control is obtained
in 60–80% with acceptable toxicity, mostly gastrointestinal and urogenital toxicity [
13
].
The current consensus is that a cumulative dose of 80 Gy in the target volume should
be reached in order to achieve >90% local control [
23
,
39
]. MRI-guided brachytherapy
techniques are required to safely reach such doses with minimizing the dose to the organs
at risk, and, in many cases, laparoscopic guidance during applicator placement is needed
Cancers 2021,13, 6275 5 of 14
to reach adequate target coverage and avoid bowel perforation [
40
]. Isolated vaginal
vault recurrences after previous postoperative vaginal vault brachytherapy can be treated
likewise, without dose-adjustment.
3.2. Pelvic Recurrence
Local control is worse for patients with a pelvic recurrence as opposed to a vaginal
recurrence with reported 5-year local control rates between 30 and 60% [
41
,
42
]. In addition,
the overall survival is better for patients with a vaginal versus pelvic nodal recurrence,
respectively, 73% and 8–14% [41].
Patients with a pelvic recurrence can be treated with EBRT with a boost to the macro-
scopic lesions. In these cases, a combination of radiation and chemotherapy or surgery
might be beneficial as well, and this needs to be individualized [16,41].
In previously irradiated patients, the incidence of locoregional recurrence is lower, but
treatment of a recurrence is more challenging. The achievable dose will be considerably
lower in such cases and mainly depends on the tolerance of the nearby organs at risk,
particularly the bowel [
23
]. Re-irradiation has long been controversial because of the high
incidence of toxicity (fistula, bowel/bladder toxicity) [
43
]. However, with the improvement
of techniques integrating imaging during radiotherapy, this might be overcome in selected
cases. Nowadays, re-irradiation with stereotactic irradiation on a conventional linac or
MRLinac (MRL) is feasible and with acceptable toxicity [44,45].
3.3. Oligometastases
Recent developments in stereotactic radiotherapy have facilitated safe irradiation of
several malignant lesions to an ablative dose. Up to five lesions are considered ‘oligometastatic
disease’, and the SABR-COMET study has demonstrated a prolonged disease free- (DFS)
and overall survival (OS) after stereotactic radiotherapy compared to standard palliative
radiotherapy in patients with a variety of primary tumors [
46
]. Scarce retrospective data
appear to confirm these results for gynecological cancer patients [47].
3.4. MR Linac (MRL)
Stereotactic body radiotherapy (SBRT) is a method of EBRT that accurately delivers a
high dose of irradiation in one or a few treatment fractions to an extracranial target [
48
].
To safely deliver these high doses, image guidance is necessary. In the case of lung or
bone metastases, cone-beam CTs (CBCT) will be sufficient for adequate image guidance.
Hence, most modern LINACS equipped with CBCT can deliver SBRT for these disease
sites. CBCTs are not sufficient to properly visualize malignant lesions in the abdomen and
viscera, such as nodal and liver metastases.
MR guided stereotactic radiotherapy, as is performed on an MRL, is now emerging
as a treatment modality for SBRT in body sites that were recently too difficult to visualize
on CT. The MR LINACS are integrated imaging and radiation systems, which enables
visualization of the target immediately before, during, and after irradiation. The irradiation
of targets very close to sensitive organs, such as the small bowel, employment of smaller
margins, and adjustment of the treatment to the anatomy on a daily basis, are now possible.
Abdominal organs tend to move, dependent on bladder or bowel filling, and daily plan
adjustment allows the radiation oncologist to give a high dose while still sparing the organs
at risk. This feature makes MR LINAC-based SBRT very appropriate for re-irradiation of
small recurrences in previously irradiated areas, such as nodal recurrences in the abdomen.
Currently, experience is still limited, but centers worldwide are gathering evidence about
the effectiveness of MR-based SBRT [49].
In summary, the treatment of locoregional recurrence can be either with (chemo)radiotherapy
or by surgical resection and is dependent on whether or not previous pelvic radiotherapy has
been applied. In Figure 2, possible flowcharts for recurrent endometrial cancer in previ-
ously irradiated patients (Figure 2A) and in patients who did not undergo previous pelvic
radiotherapy (Figure 2B) are shown. The balance between potential toxicity and benefit
Cancers 2021,13, 6275 6 of 14
should be discussed with each individual patient. In the case of oligometastatic disease,
local treatment might be of benefit.
Figure 2.
Flow chart for possible treatment decisions in patients with recurrent endometrial cancer after initial surgical
treatment with (
A
) or without (
B
) previous adjuvant locoregional radiotherapy. EBRT: external beam radiotherapy; SBRT:
stereotactic body radiotherapy. * excluding adjuvant brachytherapy only.
4. Systemic Treatment
4.1. Chemotherapy
In the chemotherapy trials for recurrent endometrial cancer, almost all the randomized
studies also included patients with locoregionally advanced endometrial cancers, which
might result in more favorable results compared to those patients with metastatic disease.
The publication of randomized trials concerning systemic chemotherapy for metastatic
endometrial cancer started in the late previous century with doxorubicin(A), later combined
with cisplatin (P) and paclitaxel (T) [
50
53
]. TAP was, for a long period, the most effective
evidence-based therapy with a significantly higher response rate of 57% versus 34% for
AP (P < 0.01), and improved PFS (median, 8.3 v 5.3 months; P < 0.01), and OS (median,
15.3 v 12.3 months; P = 0.037). However, toxicity, and especially neurological toxicity, as
well as cardiac toxicity, were a major concern in this elderly population, and many centers
started to use carboplatin and paclitaxel instead, with similar results. In 2020, the long-
awaited randomized non-inferiority study GOG0209 comparing carboplatin and paclitaxel
with paclitaxel, doxorubicin, and cisplatin confirmed that carboplatin and paclitaxel is not
inferior to TAP [54].
Only a few randomized phase III and phase II chemotherapy trials have been pub-
lished since, investigating schedules for a second recurrence, albeit without startling effects.
McMeekin et al., published an early stopped, phase III trial of ixabepilone versus either
paclitaxel or doxorubicin for second-line treatment [
55
]. Docetaxel combined with plat-
inum did not seem superior to paclitaxel [
56
,
57
], and neither was vinorelbine combined
with cisplatin [
58
], nor topotecan [
59
], dactinomycin [
60
], pegylated doxorubicin [
61
,
62
],
oxaliplatin [
63
], pemetrexed [
64
], trabectedin [
65
], or gemcitabine [
66
]. A weekly schedule
with carboplatin and paclitaxel according to a phase II trial seems to have one of the more
favorable results for patients previously treated with chemotherapy with a response rate
of 39% and a median PFS of 8 months and an OS of 9 months at the cost of increased
myelodepression and neuropathy [67].
4.2. Immunotherapy
Immunotherapy with checkpoint inhibitors, both PD1 and PDL1 inhibitors, is, nowa-
days, the most promising therapy for endometrial cancer. Two drugs are currently ap-
proved by the EMA and/or FDA; pembrolizumab and dostarlimab. Dostarlimab (Jemperli)
was granted accelerated approval both by the FDA and EMA for the treatment of patients
with recurrent or advanced deficient mismatch repair (dMMR) endometrial cancer that has
progressed or following prior treatment with platinum-containing chemotherapy [
68
]. In
patients with previously treated metastatic endometrial cancer irrespective of MSI/MMR
Cancers 2021,13, 6275 7 of 14
status, a response rate of 64% in MSI-H/MMRD and 36% for MSS/pMMR was seen with the
combination of pembrolizumab and lenvatinib [
69
]. Based on this study, pembrolizumab
with lenvatinib was also approved in an accelerated manner by the FDA for patients with
previously treated metastatic endometrial cancer whose tumors were not MSI-H/dMMR.
This study was followed by KEYNOTE-775/Study 309, a randomized phase III trial for
endometrial cancer patients with tumors that are not deficient mismatch repair or MSI high
and who have recurrent disease following prior systemic therapy. For this combination of
lenvatinib and pembrolizumab, the median OS improved from 12 months to 17.4 months,
with an HR 0.68 (95% CI 0.56–0.84) (Makker (abstract SGO 2021). The marketing authoriza-
tion application is currently under review by the EMA. The results from other Checkpoint
inhibitor phase III trials with, for example, durvalumab with olaparib (NCT 04269200) and
atezolizumab with carboplatin and paclitaxel (NCT 03603184), will become available in the
future years.
4.3. Targeted Therapy
A wide range of targeted therapies have been explored for metastatic endometrial cancers.
In the early years when these drugs became available, they were used in unselected patients;
more recently, due to increased availability of genome sequencing, more studies are focused
on specific genetic alterations, such as mutations and copy number changes in the tumor. No
randomized phase III trials have been published for targeted therapies in this patient population.
Neither has any targeted drug been approved by the FDA or EMA at this moment. A large
number of randomized phase II and phase III trials have been published on angiogene-
sis inhibitors, such as bevacizumab [
70
72
], brivanib [
73
], nintedanib [
74
], sunitinib [
75
],
cediranib [
76
], trebananib [
77
] and lenvatinib [
78
], thalidomide [
79
], and aflibercept [
80
].
Another group of drugs of interest in endometrial cancers are the mTOR inhibitors: tem-
sirolimus [
71
,
81
84
], ridaforolimus [
85
87
], and everolimus [
88
,
89
] again without success
for registration. The newer drugs, such as PIK3CA inhibitors and AKT inhibitors, have
been tested in few phase II trials, with only one looking at a PIK3CA mutation upfront [
90
]
In addition, several EGFR inhibitors were used in phase II trials, such as gefitinib [
91
] and
erlotinib [
92
]. The MEK inhibitor selumetinib also did not pass to a phase III trial [
93
].
Selinexor, an exportin 1 inhibitor, showed interesting results in endometrial cancer, and
this is one of the rare targeted drugs proceeding to a phase III trial. It is currently being
tested as maintenance after a response to carboplatin paclitaxel (NCT03555422).
5. Hormonal Treatment
Hormonal therapy for endometrial cancer has been used since the 1950s after it became
clear that progesterone could induce the regression of endometrial hyperplasia and EC [
94
].
In the first publication by Kelley and Baker in 1961, a response rate of 29% to progestin
therapy was reported among 21 recurrent ECs. This led to widespread application of
progestin therapy in clinical practice. Initially, several investigations in patients with
advanced-stage and recurrent EC confirmed the original reported response rates, with
some studies observing a response rate as high as 56% of patients [
95
,
96
]. However, recent
studies with a more modern trial design and more stringent endpoints reported a lower
response rate ranging from 11% to 24%, although patients that did respond often had
long progression-free intervals [
97
99
]. Newer hormonal drugs, such as tamoxifen and
aromatase inhibitors, have shown lower response rates than progestins and are, therefore,
regarded as second-line hormonal therapies [100,101].
The response rates are significantly higher in ER/PR positive EC [
102
,
103
]. Neverthe-
less, a recent meta-analysis showed that ER/PR status was integrated in only 70 out of 1837
included cases, indicating the limited available research on ER/PR status and response to
hormonal therapy in EC. Van Weelden et al. demonstrated that the classification of ER/PR
expression into three risk groups (0–10% (high), 20–80% (intermediate), and 90–100% (low))
resulted in better prognostication in EC, suggesting tissue specific cut-off [
104
]. Multiple
studies have shown that, during cancer progression, the loss of ER/PR occurs in at least
Cancers 2021,13, 6275 8 of 14
20% of metastasis from ER/PR positive primary tumors; this underlines the relevance of
reassessing ER/PR prior to the start of the treatment of recurrent EC [105].
Yet, the presence of ER/PR is not inherently reflecting active intracellular ER sig-
naling and hormone driven tumor growth. Therefore, the ER pathway activity testing
that indicates an activated ER signaling might improve the prediction of the response to
hormonal therapy in EC. In EC, ER pathway activity scores (ERPAS) were demonstrated
to better predict the prognosis compared to ER expression [
106
]. In a recently published
paper, pretreatment biopsies of patients with recurrent (n = 51) and advanced (n = 30)
endometrial carcinoma were analyzed for ER/PR expression and integrated the ERPAS
analysis. Interestingly, all the responders, i.e., complete- and partial response (CR, PR),
had ER/PR expression >50%. Among progestin users, the response rate (RR) was 37.7%
for ER > 50%, 56.8% for PR > 50%, and 62.1% if activated ERPAS [
107
]. In a multivariable
regression analysis, including tumor grade, histology, ER/PR, and ERPAS, the ERPAS >15
was the sole marker that remained significantly associated with PFS (HR 4.525, 95%-CI
1.85–11.07, p= 0.001). A multivariable regression analysis without ERPAS showed that PR
expression was the only variable with significant association with PFS (HR 2.964, 95%-CI
1.58–5.58, p= 0.001). In those who responded to hormonal therapy, 34.3% of the patients
with PR >50% had not progressed after 2 years.
In conclusion, patients with systemic recurrent endometrial cancer can be treated with
hormonal therapy, immunotherapy, or chemotherapy. The evidence on targeted therapy is
limited. Figure 3summarizes the systemic treatment options with response rates and data
on progression-free survival and overall survival.
Figure 3.
Overview of systemic treatment options in recurrent endometrial cancer. ER: estrogen
receptor; PR: progesterone receptor; MMR-D: mismatch repair deficient; MSI: microsatellite instability;
MSS: microsatellite stable; pMMR: proficient mismatch repair; PFS: progression-free survival; OS:
overall survival. * Preferably histology on recurrent tumor ** also approved for pMMR/MSS ***
Dependent on level of expression.
6. Conclusions and Future Perspectives
The treatment of recurrent endometrial cancer is a therapeutic challenge, especially in
the previously irradiated patient or in the patient with oligometastatic disease. In the last
decade, the improved selection of patients with recurrent endometrial cancer resulted in an
improved 5-year survival rate from 25% up to 75%. The treatment modalities can be either
local (surgery and radiotherapy) or systemic (chemotherapy, targeted therapy, hormonal
therapy, or immunotherapy). In the case of systemic therapy, evidence is available for
Cancers 2021,13, 6275 9 of 14
chemotherapy, immunotherapy, and hormonal therapy. In the case of targeted therapy, so
far, no phase III trials are available, hampering specific recommendations. It is expected
that molecular profiling in endometrial cancer will be directive not only in the adjuvant
setting but also in patients with recurrent disease [
108
]. Furthermore, combinations of
local and systemic treatment might benefit selected patients. Trials with combinations of
radiotherapy and immunotherapy or combinations of different systemic treatments are on-
going, and, hopefully, more evidence will become available in the following years. To gain
better evidence regarding the different treatment strategies, further studies are warranted.
Furthermore, to better select patients, research in the field of predictive biomarkers and the
prospective analysis of outcome in large databases is necessary.
Close collaboration between the radiation oncologist, medical oncologist, pathologist,
radiologist, and gynecologic surgeon is essential to obtain the best possible outcome for
these complex patients, and discussing these patients in a multidisciplinary tumor board
with experience in treating recurrent endometrial cancer is mandatory.
Author Contributions:
Conceptualization, J.M.A.P. and H.R., Writing original draft preparation,
H.R., C.V., W.J.v.W., A.S., J.D., N.O., J.M.A.P., Writing review and editing J.M.A.P. and H.R. All
authors have read and agreed to the published version of the manuscript.
Funding: This research received no external funding.
Conflicts of Interest: The authors declare no conflict of interest.
References
1.
Lortet-Tieulent, J.; Ferlay, J.; Bray, F.; Jemal, A. International patterns and trends in endometrial cancer incidence, 1978–2013. J.
Natl. Cancer Inst. 2018,110, 354–361. [CrossRef] [PubMed]
2. Sorosky, J.I. Endometrial cancer. Obstet. Gynecol. 2012,120, 383–397. [CrossRef] [PubMed]
3.
Onstad, M.A.; Schmandt, R.E.; Lu, K.H. Addressing the role of obesity in endometrial cancer risk, prevention, and treatment. J.
Clin. Oncol. 2016,34, 4225–4230. [CrossRef] [PubMed]
4.
Constantine, G.D.; Kessler, G.; Graham, S.; Goldstein, S.R. Increased incidence of endometrial cancer following the women’s
health initiative: An assessment of risk factors. J. Womens Health 2019,28, 237–243. [CrossRef]
5. Bokhman, J.V. Two pathogenetic types of endometrial carcinoma. Gynecol. Oncol. 1983,15, 10–17. [CrossRef]
6.
Amant, F.; Moerman, P.; Neven, P.; Timmerman, D.; Van Limbergen, E.; Vergote, I. Endometrial cancer. Lancet
2005
,366, 491–505.
[CrossRef]
7.
Kandoth, C.; Schultz, N.; Cherniack, A.D.; Akbani, R.; Liu, Y.; Shen, H.; Robertson, A.G.; Pashtan, I.; Shen, R.; Benz, C.C.; et al.
Integrated genomic characterization of endometrial carcinoma. Nature 2013,497, 67–73. [CrossRef]
8.
Yen, T.T.; Wang, T.L.; Fader, A.N.; Shih, I.M.; Gaillard, S. Molecular classification and emerging targeted therapy in endometrial
cancer. Int. J. Gynecol. Pathol. 2020,39, 26–35. [CrossRef]
9.
Huvila, J.; McAlpine, J.N. UpToDate Endometrial Cancer: Pathology and Classification. Available online: https://www.uptodate.
com/contents/endometrial-cancer-pathology-and-classification?source=history_widget#subscribeMessage (accessed on 17
September 2021).
10.
Paleari, L.; Pesce, S.; Rutigliani, M.; Greppi, M.; Obino, V.; Gorlero, F.; Vellone, V.G.; Marcenaro, E. New insights into endometrial
cancer. Cancers 2021,13, 1496. [CrossRef]
11. Morice, P.; Leary, A.; Creutzberg, C.; Abu-Rustum, N.; Darai, E. Endometrial cancer. Lancet 2016,387, 1094–1108. [CrossRef]
12.
Del Carmen, M.G.; Boruta, D.M., 2nd; Schorge, J.O. Recurrent endometrial cancer. Clin. Obstet. Gynecol.
2011
,54, 266–277.
[CrossRef]
13.
Creutzberg, C.L.; van Putten, W.L.; Koper, P.C.; Lybeert, M.L.; Jobsen, J.J.; Wárlám-Rodenhuis, C.C.; De Winter, K.A.; Lutgens,
L.C.; van den Bergh, A.C.; van der Steen-Banasik, E.; et al. Survival after relapse in patients with endometrial cancer: Results
from a randomized trial. Gynecol. Oncol. 2003,89, 201–209. [CrossRef]
14.
Legge, F.; Restaino, S.; Leone, L.; Carone, V.; Ronsini, C.; Di Fiore, G.L.M.; Pasciuto, T.; Pelligra, S.; Ciccarone, F.; Scambia, G.;
et al. Clinical outcome of recurrent endometrial cancer: Analysis of post-relapse survival by pattern of recurrence and secondary
treatment. Int. J. Gynecol. Cancer 2020,30, 193–200. [CrossRef]
15.
Creutzberg, C.L.; van Putten, W.L.; Koper, P.C.; Lybeert, M.L.; Jobsen, J.J.; Warlam-Rodenhuis, C.C.; De Winter, K.A.; Lutgens,
L.C.; van den Bergh, A.C.; van de Steen-Banasik, E.; et al. Surgery and postoperative radiotherapy versus surgery alone for
patients with stage-1 endometrial carcinoma: Multicentre randomised trial. PORTEC study group. Post operative radiation
therapy in endometrial carcinoma. Lancet 2000,355, 1404–1411. [CrossRef]
16.
Francis, S.R.; Ager, B.J.; Do, O.A.; Huang, Y.J.; Soisson, A.P.; Dodson, M.K.; Werner, T.L.; Sause, W.T.; Grant, J.D.; Gaffney, D.K.
Recurrent early stage endometrial cancer: Patterns of recurrence and results of salvage therapy. Gynecol. Oncol.
2019
,154, 38–44.
[CrossRef]
Cancers 2021,13, 6275 10 of 14
17.
Matei, D.; Filiaci, V.; Randall, M.E.; Mutch, D.; Steinhoff, M.M.; Di Silvestro, P.A.; Moxley, K.M.; Kim, Y.M.; Powell, M.A.; O’Malley,
D.M.; et al. Adjuvant chemotherapy plus radiation for locally advanced endometrial cancer. N. Engl. J. Med.
2019
,380, 2317–2326.
[CrossRef]
18.
De Boer, S.M.; Powell, M.E.; Mileshkin, L.; Katsaros, D.; Bessette, P.; Haie-Meder, C.; Ottevanger, P.B.; Ledermann, J.A.; Khaw,
P.; Colombo, A.; et al. Adjuvant chemoradiotherapy versus radiotherapy alone for women with high-risk endometrial cancer
(PORTEC-3): Final results of an international, open-label, multicentre, randomised, phase 3 trial. Lancet Oncol.
2018
,19, 295–309.
[CrossRef]
19.
Connor, E.V.; Rose, P.G. Management strategies for recurrent endometrial cancer. Expert Rev. Anticancer Ther.
2018
,18, 873–885.
[CrossRef]
20.
Van Wijk, F.H.; van der Burg, M.E.; Burger, C.W.; Vergote, I.; van Doorn, H.C. Management of recurrent endometrioid endometrial
carcinoma: An overview. Int. J. Gynecol. Cancer 2009,19, 314–320. [CrossRef]
21.
Bristow, R.E.; Santillan, A.; Zahurak, M.L.; Gardner, G.J.; Giuntoli, R.L., II; Armstrong, D.K. Salvage cytoreductive surgery for
recurrent endometrial cancer. Gynecol. Oncol. 2006,103, 281–287. [CrossRef]
22.
Wylie, J.; Irwin, C.; Pintilie, M.; Levin, W.; Manchul, L.; Milosevic, M.; Fyles, A. Results of radical radiotherapy for recurrent
endometrial cancer. Gynecol. Oncol. 2000,77, 66–72. [CrossRef]
23.
Kamrava, M.; Beriwal, S.; Erickson, B.; Gaffney, D.; Jhingran, A.; Klopp, A.; Park, S.J.; Viswanathan, A.; Yashar, C.; Lin, L.
American Brachytherapy Society recurrent carcinoma of the endometrium task force patterns of care and review of the literature.
Brachytherapy 2017,16, 1129–1143. [CrossRef]
24.
Hardarson, H.A.; Heidemann, L.N.; dePont Christensen, R.; Mogensen, O.; Jochumsen, K.M. Vaginal vault recurrences of
endometrial cancer in non-irradiated patients—Radiotherapy or surgery. Gynecol. Oncol. Rep. 2015,11, 26–30. [CrossRef]
25.
Chiantera, V.; Rossi, M.; De Iaco, P.; Koehler, C.; Marnitz, S.; Gallotta, V.; Margariti, A.P.; Parazzini, F.; Scambia, G.; Schneider, A.;
et al. Pelvic exenteration for recurrent endometrial adenocarcinoma: A retrospective multi-institutional study about 21 patients.
Int. J. Gynecol. Cancer 2014,24, 880–884. [CrossRef]
26.
Morris, M.; Alvarez, R.D.; Kinney, W.K.; Wilson, T.O. Treatment of recurrent adenocarcinoma of the endometrium with pelvic
exenteration. Gynecol. Oncol. 1996,60, 288–291. [CrossRef]
27.
Khoury-Collado, F.; Einstein, M.H.; Bochner, B.H.; Alektiar, K.M.; Sonoda, Y.; Abu-Rustum, N.R.; Brown, C.L.; Gardner, G.J.;
Barakat, R.R.; Chi, D.S. Pelvic exenteration with curative intent for recurrent uterine malignancies. Gynecol. Oncol.
2012
,124,
42–47. [CrossRef]
28.
Shikama, A.; Minaguchi, T.; Takao, W.; Hosokawa, Y.; Nishida, K.; Tasaka, N.; Akiyama, A.; Sakurai, M.; Ochi, H.; Satoh, T.
Predictors of favorable survival after secondary cytoreductive surgery for recurrent endometrial cancer. Int. J. Clin. Oncol.
2019
,
24, 1256–1263. [CrossRef]
29.
Turan, T.; Tasci, T.; Karalok, A.; Ureyen, I.; Kocak, O.; Turkmen, O.; Basaran, D.; Tulunay, G. Salvage cytoreductive surgery for
recurrent endometrial cancer. Int. J. Gynecol. Cancer 2015,25, 1623–1632. [CrossRef]
30.
Papadia, A.; Bellati, F.; Ditto, A.; Bogani, G.; Gasparri, M.L.; Di Donato, V.; Martinelli, F.; Lorusso, D.; Benedetti-Panici, P.;
Raspagliesi, F. Surgical treatment of recurrent endometrial cancer: Time for a paradigm shift. Ann. Surg. Oncol.
2015
,22,
4204–4210. [CrossRef]
31.
Germanova, A.; Raspagliesi, F.; Chiva, L.; Dusek, L.; Arvas, M.; Leblanc, E.; Lengeyl, T.; Di Donato, V.; Zaal, A.; Dursun, P.;
et al. Oncological outcome of surgical management in patients with recurrent uterine cancer-a multicenter retrospective cohort
study-CEEGOG EX01 trial. Int. J. Gynecol. Cancer 2019,29, 711–720. [CrossRef]
32.
Ren, Y.; Shan, B.; Shi, D.; Wang, H. Salvage cytoreductive surgery for patients with recurrent endometrial cancer: A retrospective
study. BMC Cancer 2014,14, 135. [CrossRef] [PubMed]
33.
Awtrey, C.S.; Cadungog, M.G.; Leitao, M.M.; Alektiar, K.M.; Aghajanian, C.; Hummer, A.J.; Barakat, R.R.; Chi, D.S. Surgical
resection of recurrent endometrial carcinoma. Gynecol. Oncol. 2006,102, 480–488. [CrossRef] [PubMed]
34.
Campagnutta, E.; Giorda, G.; De Piero, G.; Sopracordevole, F.; Visentin, M.C.; Martella, L.; Scarabelli, C. Surgical treatment of
recurrent endometrial carcinoma. Cancer 2004,100, 89–96. [CrossRef] [PubMed]
35.
Barlin, J.N.; Puri, I.; Bristow, R.E. Cytoreductive surgery for advanced or recurrent endometrial cancer: A meta-analysis. Gynecol.
Oncol. 2010,118, 14–18. [CrossRef] [PubMed]
36.
Vandenput, I.; Van Calster, B.; Capoen, A.; Leunen, K.; Berteloot, P.; Neven, P.; Moerman, P.; Vergote, I.; Amant, F. Neoadjuvant
chemotherapy followed by interval debulking surgery in patients with serous endometrial cancer with transperitoneal spread
(stage IV): A new preferred treatment? Br. J. Cancer 2009,101, 244–249. [CrossRef] [PubMed]
37.
Piura, B.; Rabinovich, A.; Apel-Sarid, L.; Shaco-Levy, R. Splenic metastasis from endometrial carcinoma: Report of a case and
review of literature. Arch. Gynecol. Obstet. 2009,280, 1001–1006. [CrossRef] [PubMed]
38.
Tangjitgamol, S.; Levenback, C.F.; Beller, U.; Kavanagh, J.J. Role of surgical resection for lung, liver, and central nervous system
metastases in patients with gynecological cancer: A literature review. Int. J. Gynecol. Cancer
2004
,14, 399–422. [CrossRef]
[PubMed]
39.
Sapienza, L.G.; Ning, M.S.; de la Pena, R.; McNew, L.K.; Jhingran, A.; Georgeon, L.; Rasool, N.; Gomes, M.J.L.; Abu-Isa, E.;
Baiocchi, G. Outcomes and toxicity after salvage radiotherapy for vaginal relapse of endometrial cancer. Int. J. Gynecol. Cancer
2020,30, 1535–1541. [CrossRef]
Cancers 2021,13, 6275 11 of 14
40.
Fokdal, L.; Tanderup, K.; Nielsen, S.K.; Christensen, H.K.; Røhl, L.; Pedersen, E.M.; Schønemann, N.K.; Lindegaard, J.C. Image
and laparoscopic guided interstitial brachytherapy for locally advanced primary or recurrent gynaecological cancer using the
adaptive GEC ESTRO target concept. Radiother. Oncol. 2011,100, 473–479. [CrossRef]
41.
Ho, J.C.; Allen, P.K.; Jhingran, A.; Westin, S.N.; Lu, K.H.; Eifel, P.J.; Klopp, A.H. Management of nodal recurrences of endometrial
cancer with IMRT. Gynecol. Oncol. 2015,139, 40–46. [CrossRef]
42.
Lindemann, K.; Smogeli, E.; Småstuen, M.C.; Bruheim, K.; Trovik, J.; Nordberg, T.; Kristensen, G.B.; Werner, H.M.J.; Nakken, E.
Salvage radiation for pelvic relapse after surgically treated endometrial cancer. Cancers 2021,13, 1367. [CrossRef]
43. Llewelyn, M.; Taylor, A. Re-irradiation of cervical and endometrial cancer. Curr. Opin. Oncol. 2017,29, 343–350. [CrossRef]
44.
Guckenberger, M.; Bachmann, J.; Wulf, J.; Mueller, G.; Krieger, T.; Baier, K.; Richter, A.; Wilbert, J.; Flentje, M. Stereotactic body
radiotherapy for local boost irradiation in unfavourable locally recurrent gynaecological cancer. Radiother. Oncol.
2010
,94, 53–59.
[CrossRef]
45.
Higginson, D.S.; Morris, D.E.; Jones, E.L.; Clarke-Pearson, D.; Varia, M.A. Stereotactic body radiotherapy (SBRT): Technological
innovation and application in gynecologic oncology. Gynecol. Oncol. 2011,120, 404–412. [CrossRef]
46.
Palma, D.A.; Olson, R.; Harrow, S.; Gaede, S.; Louie, A.V.; Haasbeek, C.; Mulroy, L.; Lock, M.; Rodrigues, G.B.; Yaremko, B.P.;
et al. Stereotactic ablative radiotherapy for the comprehensive treatment of oligometastatic cancers: Long-term results of the
SABR-COMET phase II randomized trial. J. Clin. Oncol. 2020,38, 2830–2838. [CrossRef]
47.
Ning, M.S.; Ahobila, V.; Jhingran, A.; Stecklein, S.R.; Frumovitz, M.; Schmeler, K.M.; Eifel, P.J.; Klopp, A.H. Outcomes and patterns
of relapse after definitive radiation therapy for oligometastatic cervical cancer. Gynecol. Oncol. 2018,148, 132–138. [CrossRef]
48.
Guckenberger, M.; Andratschke, N.; Alheit, H.; Holy, R.; Moustakis, C.; Nestle, U.; Sauer, O. Definition of stereotactic body
radiotherapy: Principles and practice for the treatment of stage I non-small cell lung cancer. Strahlenther. Onkol.
2014
,190, 26–33.
[CrossRef]
49.
De Mol van Otterloo, S.R.; Christodouleas, J.P.; Blezer, E.L.A.; Akhiat, H.; Brown, K.; Choudhury, A.; Eggert, D.; Erickson, B.A.;
Faivre-Finn, C.; Fuller, C.D.; et al. The MOMENTUM study: An international registry for the evidence-based introduction of
MR-guided adaptive therapy. Front. Oncol. 2020,10, 1328. [CrossRef]
50.
Thigpen, J.T.; Blessing, J.A.; DiSaia, P.J.; Yordan, E.; Carson, L.F.; Evers, C. A randomized comparison of doxorubicin alone
versus doxorubicin plus cyclophosphamide in the management of advanced or recurrent endometrial carcinoma: A gynecologic
oncology group study. J. Clin. Oncol. 1994,12, 1408–1414. [CrossRef]
51.
Van Wijk, F.H.; Aapro, M.S.; Bolis, G.; Chevallier, B.; van der Burg, M.E.; Poveda, A.; de Oliveira, C.F.; Tumolo, S.; Scotto di
Palumbo, V.; Piccart, M.; et al. Doxorubicin versus doxorubicin and cisplatin in endometrial carcinoma: Definitive results of a
randomised study (55872) by the EORTC gynaecological cancer group. Ann. Oncol. 2003,14, 441–448. [CrossRef]
52.
Gallion, H.H.; Brunetto, V.L.; Cibull, M.; Lentz, S.S.; Reid, G.; Soper, J.T.; Burger, R.A.; Andersen, W. Randomized phase III trial
of standard timed doxorubicin plus cisplatin versus circadian timed doxorubicin plus cisplatin in stage III and IV or recurrent
endometrial carcinoma: A gynecologic oncology group study. J. Clin. Oncol. 2003,21, 3808–3813. [CrossRef]
53.
Fleming, G.F.; Brunetto, V.L.; Cella, D.; Look, K.Y.; Reid, G.C.; Munkarah, A.R.; Kline, R.; Burger, R.A.; Goodman, A.; Burks, R.T.
Phase III trial of doxorubicin plus cisplatin with or without paclitaxel plus filgrastim in advanced endometrial carcinoma: A
gynecologic oncology group study. J. Clin. Oncol. 2004,22, 2159–2166. [CrossRef]
54.
Miller, D.S.; Filiaci, V.L.; Mannel, R.S.; Cohn, D.E.; Matsumoto, T.; Tewari, K.S.; DiSilvestro, P.; Pearl, M.L.; Argenta, P.A.; Powell,
M.A.; et al. Carboplatin and paclitaxel for advanced endometrial cancer: Final overall survival and adverse event analysis of a
phase III trial (NRG Oncology/GOG0209). J. Clin. Oncol. 2020,38, 3841–3850. [CrossRef]
55.
McMeekin, D.S.; Sill, M.W.; Walker, J.L.; Moore, K.N.; Waggoner, S.E.; Thaker, P.H.; Rizack, T.; Hoffman, J.S.; Fracasso, P.M. A
phase I study of IV doxorubicin plus intraperitoneal (IP) paclitaxel and IV or IP cisplatin in endometrial cancer patients at high
risk for peritoneal failure (GOG 9920): An NRG oncology/gynecologic oncology group study. Gynecol. Oncol.
2015
,138, 36–40.
[CrossRef]
56.
Garcia, A.A.; Blessing, J.A.; Nolte, S.; Mannel, R.S. A phase II evaluation of weekly docetaxel in the treatment of recurrent or
persistent endometrial carcinoma: A study by the gynecologic oncology group. Gynecol. Oncol. 2008,111, 22–26. [CrossRef]
57.
Nomura, H.; Aoki, D.; Takahashi, F.; Katsumata, N.; Watanabe, Y.; Konishi, I.; Jobo, T.; Hatae, M.; Hiura, M.; Yaegashi, N.
Randomized phase II study comparing docetaxel plus cisplatin, docetaxel plus carboplatin, and paclitaxel plus carboplatin in
patients with advanced or recurrent endometrial carcinoma: A Japanese gynecologic oncology group study (JGOG2041). Ann.
Oncol. 2011,22, 636–642. [CrossRef]
58.
Gebbia, V.; Testa, A.; Borsellino, N.; Ferrera, P.; Tirrito, M.; Palmeri, S. Cisplatin and vinorelbine in advanced and/or metastatic
adenocarcinoma of the endometrium: A new highly active chemotherapeutic regimen. Ann. Oncol.
2001
,12, 767–772. [CrossRef]
59.
Miller, D.S.; Blessing, J.A.; Lentz, S.S.; Waggoner, S.E. A phase II trial of topotecan in patients with advanced, persistent, or
recurrent endometrial carcinoma: A gynecologic oncology group study. Gynecol. Oncol. 2002,87, 247–251. [CrossRef]
60.
Moore, D.H.; Blessing, J.A.; Dunton, C.; Buller, R.E.; Reid, G.C. Dactinomycin in the treatment of recurrent or persistent
endometrial carcinoma: A phase II study of the gynecologic oncology group. Gynecol. Oncol. 1999,75, 473–475. [CrossRef]
61.
Muggia, F.M.; Blessing, J.A.; Sorosky, J.; Reid, G.C. Phase II trial of the pegylated liposomal doxorubicin in previously treated
metastatic endometrial cancer: A gynecologic oncology group study. J. Clin. Oncol. 2002,20, 2360–2364. [CrossRef]
62.
Escobar, P.F.; Markman, M.; Zanotti, K.; Webster, K.; Belinson, J. Phase 2 trial of pegylated liposomal doxorubicin in advanced
endometrial cancer. J. Cancer Res. Clin. Oncol. 2003,129, 651–654. [CrossRef] [PubMed]
Cancers 2021,13, 6275 12 of 14
63.
Fracasso, P.M.; Blessing, J.A.; Molpus, K.L.; Adler, L.M.; Sorosky, J.I.; Rose, P.G. Phase II study of oxaliplatin as second-line
chemotherapy in endometrial carcinoma: A gynecologic oncology group study. Gynecol. Oncol.
2006
,103, 523–526. [CrossRef]
[PubMed]
64.
Miller, D.S.; Blessing, J.A.; Drake, R.D.; Higgins, R.; McMeekin, D.S.; Puneky, L.V.; Krasner, C.N. A phase II evaluation of
pemetrexed (Alimta, LY231514, IND #40061) in the treatment of recurrent or persistent endometrial carcinoma: A phase II study
of the gynecologic oncology. Gynecol. Oncol. 2009,115, 443–446. [CrossRef]
65.
McMeekin, D.S.; Lisyanskaya, A.; Crispens, M.; Oza, A.M.; Braly, P.; Doering, D.; Bayever, E.; Michiels, B.; Markman, M.
Single-agent trabectedin as second-line therapy of persistent or recurrent endometrial cancer: Results of a multicenter phase II
study. Gynecol. Oncol. 2009,114, 288–292. [CrossRef] [PubMed]
66.
Tait, D.L.; Blessing, J.A.; Hoffman, J.S.; Moore, K.N.; Spirtos, N.M.; Lachance, J.A.; Rotmensch, J.; Miller, D.S. A phase II study
of gemcitabine (gemzar, LY188011) in the treatment of recurrent or persistent endometrial carcinoma: A gynecologic oncology
group study. Gynecol. Oncol. 2011,121, 118–121. [CrossRef]
67.
Vandenput, I.; Vergote, I.; Neven, P.; Amant, F. Weekly paclitaxel-carboplatin regimen in patients with primary advanced or
recurrent endometrial carcinoma. Int. J. Gynecol. Cancer 2012,22, 617–622. [CrossRef]
68.
Oaknin, A.; Tinker, A.V.; Gilbert, L.; Samouëlian, V.; Mathews, C.; Brown, J.; Barretina-Ginesta, M.P.; Moreno, V.; Gravina, A.;
Abdeddaim, C.; 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, 1766–1772. [CrossRef]
69.
Makker, V.; Taylor, M.H.; Aghajanian, C.; Oaknin, A.; Mier, J.; Cohn, A.L.; Romeo, M.; Bratos, R.; Brose, M.S.; DiSimone, C.; et al.
Lenvatinib plus pembrolizumab in patients with advanced endometrial cancer. J. Clin. Oncol. 2020,38, 2981–2992. [CrossRef]
70.
Lorusso, D.; Ferrandina, G.; Colombo, N.; Pignata, S.; Pietragalla, A.; Sonetto, C.; Pisano, C.; Lapresa, M.T.; Savarese, A.;
Tagliaferri, P.; 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, 406–412. [CrossRef]
71.
Aghajanian, C.; Filiaci, V.; Dizon, D.S.; Carlson, J.W.; Powell, M.A.; Secord, A.A.; Tewari, K.S.; Bender, D.P.; O’Malley, D.M.;
Stuckey, A.; 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, 274–281. [CrossRef]
72.
Aghajanian, C.; Sill, M.W.; Darcy, K.M.; Greer, B.; McMeekin, D.S.; Rose, P.G.; Rotmensch, J.; Barnes, M.N.; Hanjani, P.; Leslie, K.K.
Phase II trial of bevacizumab in recurrent or persistent endometrial cancer: A gynecologic oncology group study. J. Clin. Oncol.
2011,29, 2259–2265. [CrossRef]
73.
Powell, M.A.; Sill, M.W.; Goodfellow, P.J.; Benbrook, D.M.; Lankes, H.A.; Leslie, K.K.; Jeske, Y.; Mannel, R.S.; Spillman, M.A.; Lee,
P.S.; et al. A phase II trial of brivanib in recurrent or persistent endometrial cancer: An NRG oncology/gynecologic oncology
group study. Gynecol. Oncol. 2014,135, 38–43. [CrossRef]
74.
Dizon, D.S.; Sill, M.W.; Schilder, J.M.; McGonigle, K.F.; Rahman, Z.; Miller, D.S.; Mutch, D.G.; Leslie, K.K. A phase II evaluation of
nintedanib (BIBF-1120) in the treatment of recurrent or persistent endometrial cancer: An NRG oncology/gynecologic oncology
group study. Gynecol. Oncol. 2014,135, 441–445. [CrossRef]
75.
Castonguay, V.; Lheureux, S.; Welch, S.; Mackay, H.J.; Hirte, H.; Fleming, G.; Morgan, R.; Wang, L.; Blattler, C.; Ivy, P.S.; et al.
A phase II trial of sunitinib in women with metastatic or recurrent endometrial carcinoma: A study of the Princess Margaret,
Chicago and California consortia. Gynecol. Oncol. 2014,134, 274–280. [CrossRef]
76.
Bender, D.; Sill, M.W.; Lankes, H.A.; Reyes, H.D.; Darus, C.J.; Delmore, J.E.; Rotmensch, J.; Gray, H.J.; Mannel, R.S.; Schilder,
J.M.; et al. A phase II evaluation of cediranib in the treatment of recurrent or persistent endometrial cancer: An NRG oncol-
ogy/gynecologic oncology group study. Gynecol. Oncol. 2015,138, 507–512. [CrossRef]
77.
Moore, K.N.; Sill, M.W.; Tenney, M.E.; Darus, C.J.; Griffin, D.; Werner, T.L.; Rose, P.G.; Behrens, R. A phase II trial of trebananib
(AMG 386; IND#111071), a selective angiopoietin 1/2 neutralizing peptibody, in patients with persistent/recurrent carcinoma of
the endometrium: An NRG/gynecologic oncology group trial. Gynecol. Oncol. 2015,138, 513–518. [CrossRef]
78.
Vergote, I.; Powell, M.A.; Teneriello, M.G.; Miller, D.S.; Garcia, A.A.; Mikheeva, O.N.; Bidzinski, M.; Cebotaru, C.L.; Dutcus, C.E.;
Ren, M.; et al. Second-line lenvatinib in patients with recurrent endometrial cancer. Gynecol. Oncol.
2020
,156, 575–582. [CrossRef]
79.
McMeekin, D.S.; Sill, M.W.; Benbrook, D.; Darcy, K.M.; Stearns-Kurosawa, D.J.; Eaton, L.; Yamada, S.D. A phase II trial of
thalidomide in patients with refractory endometrial cancer and correlation with angiogenesis biomarkers: A gynecologic
oncology group study. Gynecol. Oncol. 2007,105, 508–516. [CrossRef]
80.
Coleman, R.L.; Sill, M.W.; Lankes, H.A.; Fader, A.N.; Finkler, N.J.; Hoffman, J.S.; Rose, P.G.; Sutton, G.P.; Drescher, C.W.;
McMeekin, D.S.; et al. A phase II evaluation of aflibercept in the treatment of recurrent or persistent endometrial cancer: A
gynecologic oncology group study. Gynecol. Oncol. 2012,127, 538–543. [CrossRef]
81.
Emons, G.; Kurzeder, C.; Schmalfeldt, B.; Neuser, P.; de Gregorio, N.; Pfisterer, J.; Park-Simon, T.W.; Mahner, S.; Schröder, W.;
Lück, H.J.; et al. Temsirolimus in women with platinum-refractory/resistant ovarian cancer or advanced/recurrent endometrial
carcinoma. A phase II study of the AGO-study group (AGO-GYN8). Gynecol. Oncol. 2016,140, 450–456. [CrossRef]
82. Fleming, G.F.; Filiaci, V.L.; Marzullo, B.; Zaino, R.J.; Davidson, S.A.; Pearl, M.; Makker, V.; Burke, J.J., 2nd; Zweizig, S.L.; Van Le,
L.; et al. Temsirolimus with or without megestrol acetate and tamoxifen for endometrial cancer: A gynecologic oncology group
study. Gynecol. Oncol. 2014,132, 585–592. [CrossRef]
Cancers 2021,13, 6275 13 of 14
83.
Alvarez, E.A.; Brady, W.E.; Walker, J.L.; Rotmensch, J.; Zhou, X.C.; Kendrick, J.E.; Yamada, S.D.; Schilder, J.M.; Cohn, D.E.;
Harrison, C.R.; et al. Phase II trial of combination bevacizumab and temsirolimus in the treatment of recurrent or persistent
endometrial carcinoma: A gynecologic oncology group study. Gynecol. Oncol. 2013,129, 22–27. [CrossRef]
84.
Oza, A.M.; Elit, L.; Tsao, M.S.; Kamel-Reid, S.; Biagi, J.; Provencher, D.M.; Gotlieb, W.H.; Hoskins, P.J.; Ghatage, P.; Tonkin, K.S.;
et al. Phase II study of temsirolimus in women with recurrent or metastatic endometrial cancer: A trial of the NCIC clinical trials
group. J. Clin. Oncol. 2011,29, 3278–3285. [CrossRef]
85.
Colombo, N.; McMeekin, D.S.; Schwartz, P.E.; Sessa, C.; Gehrig, P.A.; Holloway, R.; Braly, P.; Matei, D.; Morosky, A.; Dodion, P.F.;
et al. Ridaforolimus as a single agent in advanced endometrial cancer: Results of a single-arm, phase 2 trial. Br. J. Cancer
2013
,
108, 1021–1026. [CrossRef]
86.
Oza, A.M.; Pignata, S.; Poveda, A.; McCormack, M.; Clamp, A.; Schwartz, B.; Cheng, J.; Li, X.; Campbell, K.; Dodion, P.; et al.
Randomized phase II trial of ridaforolimus in advanced endometrial carcinoma. J. Clin. Oncol.
2015
,33, 3576–3582. [CrossRef]
[PubMed]
87.
Tsoref, D.; Welch, S.; Lau, S.; Biagi, J.; Tonkin, K.; Martin, L.A.; Ellard, S.; Ghatage, P.; Elit, L.; Mackay, H.J.; et al. Phase II study of
oral ridaforolimus in women with recurrent or metastatic endometrial cancer. Gynecol. Oncol.
2014
,135, 184–189. [CrossRef]
[PubMed]
88.
Slomovitz, B.M.; Lu, K.H.; Johnston, T.; Coleman, R.L.; Munsell, M.; Broaddus, R.R.; Walker, C.; Ramondetta, L.M.; Burke, T.W.;
Gershenson, D.M.; et al. A phase 2 study of the oral mammalian target of rapamycin inhibitor, everolimus, in patients with
recurrent endometrial carcinoma. Cancer 2010,116, 5415–5419. [CrossRef] [PubMed]
89.
Ray-Coquard, I.; Favier, L.; Weber, B.; Roemer-Becuwe, C.; Bougnoux, P.; Fabbro, M.; Floquet, A.; Joly, F.; Plantade, A.; Paraiso, D.;
et al. Everolimus as second- or third-line treatment of advanced endometrial cancer: ENDORAD, a phase II trial of GINECO. Br.
J. Cancer 2013,108, 1771–1777. [CrossRef] [PubMed]
90.
Myers, A.P.; Konstantinopoulos, P.A.; Barry, W.T.; Luo, W.; Broaddus, R.R.; Makker, V.; Drapkin, R.; Liu, J.; Doyle, A.; Horowitz,
N.S.; et al. Phase II, 2-stage, 2-arm, PIK3CA mutation stratified trial of MK-2206 in recurrent endometrial cancer. Int. J. Cancer
2020,147, 413–422. [CrossRef]
91.
Leslie, K.K.; Sill, M.W.; Fischer, E.; Darcy, K.M.; Mannel, R.S.; Tewari, K.S.; Hanjani, P.; Wilken, J.A.; Baron, A.T.; Godwin, A.K.;
et al. A phase II evaluation of gefitinib in the treatment of persistent or recurrent endometrial cancer: A gynecologic oncology
group study. Gynecol. Oncol. 2013,129, 486–494. [CrossRef]
92.
Oza, A.M.; Eisenhauer, E.A.; Elit, L.; Cutz, J.C.; Sakurada, A.; Tsao, M.S.; Hoskins, P.J.; Biagi, J.; Ghatage, P.; Mazurka, J.; et al.
Phase II study of erlotinib in recurrent or metastatic endometrial cancer: NCIC IND-148. J. Clin. Oncol.
2008
,26, 4319–4325.
[CrossRef]
93.
Coleman, R.L.; Sill, M.W.; Thaker, P.H.; Bender, D.P.; Street, D.; McGuire, W.P.; Johnston, C.M.; Rotmensch, J. A phase II evaluation
of selumetinib (AZD6244, ARRY-142886), a selective MEK-1/2 inhibitor in the treatment of recurrent or persistent endometrial
cancer: An NRG oncology/gynecologic oncology group study. Gynecol. Oncol. 2015,138, 30–35. [CrossRef]
94.
Kelley, R.M.; Baker, W.H. Progestational agents in the treatment of carcinoma of the endometrium. N. Engl. J. Med.
1961
,264,
216–222. [CrossRef]
95.
Bonte, J.; Decoster, J.M.; Ide, P.; Billiet, G. Hormonoprophylaxis and hormonotherapy in the treatment of endometrial adenocarci-
noma by means of medroxyprogesterone acetate. Gynecol. Oncol. 1978,6, 60–75. [CrossRef]
96.
Waterman, E.A.; Benson, R.C. Medrogestone therapy in advanced endometrial adenocarcinoma. Obstet. Gynecol.
1967
,30,
626–634.
97.
Thigpen, J.T.; Brady, M.F.; Alvarez, R.D.; Adelson, M.D.; Homesley, H.D.; Manetta, A.; Soper, J.T.; Given, F.T. Oral medroxypro-
gesterone acetate in the treatment of advanced or recurrent endometrial carcinoma: A dose-response study by the gynecologic
oncology group. J. Clin. Oncol. 1999,17, 1736–1744. [CrossRef]
98.
Lentz, S.S.; Brady, M.F.; Major, F.J.; Reid, G.C.; Soper, J.T. High-dose megestrol acetate in advanced or recurrent endometrial
carcinoma: A gynecologic oncology group study. J. Clin. Oncol. 1996,14, 357–361. [CrossRef]
99.
Podratz, K.C.; O’Brien, P.C.; Malkasian, G.D., Jr.; Decker, D.G.; Jefferies, J.A.; Edmonson, J.H. Effects of progestational agents in
treatment of endometrial carcinoma. Obstet. Gynecol. 1985,66, 106–110.
100.
Mileshkin, L.; Edmondson, R.; O’Connell, R.L.; Sjoquist, K.M.; Andrews, J.; Jyothirmayi, R.; Beale, P.; Bonaventura, T.; Goh,
J.; Hall, M.; et al. Phase 2 study of anastrozole in recurrent estrogen (ER)/progesterone (PR) positive endometrial cancer: The
PARAGON trial—ANZGOG 0903. Gynecol. Oncol. 2019,154, 29–37. [CrossRef]
101.
Lindemann, K.; Malander, S.; Christensen, R.D.; Mirza, M.R.; Kristensen, G.B.; Aavall-Lundqvist, E.; Vergote, I.; Rosenberg, P.;
Boman, K.; Nordstrøm, B. Examestane in advanced or recurrent endometrial carcinoma: A prospective phase II study by the
Nordic Society of Gynecologic Oncology (NSGO). BMC Cancer 2014,14, 68. [CrossRef]
102. Decruze, S.B.; Green, J.A. Hormone therapy in advanced and recurrent endometrial cancer: A systematic review. Int. J. Gynecol.
Cancer 2007,17, 964–978. [CrossRef]
103.
Ethier, J.L.; Desautels, D.N.; Amir, E.; MacKay, H. Is hormonal therapy effective in advanced endometrial cancer? A systematic
review and meta-analysis. Gynecol. Oncol. 2017,147, 158–166. [CrossRef]
104.
Van Weelden, W.J.; Reijnen, C.; Küsters-Vandevelde, H.V.N.; Bulten, J.; Bult, P.; Leung, S.; Visser, N.C.M.; Santacana, M.; Bronsert,
P.; Hirschfeld, M.; et al. The cutoff for estrogen and progesterone receptor expression in endometrial cancer revisited: A European
Network for Individualized Treatment of Endometrial Cancer collaboration study. Hum. Pathol. 2021,109, 80–91. [CrossRef]
Cancers 2021,13, 6275 14 of 14
105.
Concin, N.; Matias-Guiu, X.; Vergote, I.; Cibula, D.; Mirza, M.R.; Marnitz, S.; Ledermann, J.; Bosse, T.; Chargari, C.; Fagotti, A.;
et al. ESGO/ESTRO/ESP guidelines for the management of patients with endometrial carcinoma. Int. J. Gynecol. Cancer
2021
,31,
12–39. [CrossRef]
106.
Van Weelden, W.J.; van der Putten, L.J.M.; Inda, M.A.; van Brussel, A.; Snijders, M.; Schriever, L.M.M.; Bulten, J.; Massuger, L.;
van de Stolpe, A.; Pijnenborg, J.M.A. Oestrogen receptor pathway activity is associated with outcome in endometrial cancer. Br. J.
Cancer 2020,123, 785–792. [CrossRef]
107.
Van Weelden, W.J.; Lalisang, R.I.; Bulten, J.; Lindemann, K.; van Beekhuizen, H.J.; Trum, H.; Boll, D.; Werner, H.M.J.; van
Lonkhuijzen, L.R.C.W.; Yigit, R.; et al. Impact of hormonal biomarkers on response to hormonal therapy in advanced and
recurrent endometrial Cancer. Am. J. Obstet. Gynecol. 2021. [CrossRef]
108.
Urick, M.E.; Bell, D.W. Clinical actionability of molecular targets in endometrial cancer. Nat. Rev. Cancer
2019
,19, 510–521.
[CrossRef]
... In a 2006 review, Fung-Kee-Fung et al. [28] described a recurrence rate of 13% for EC of all risk stratifications. Other authors describe rates of 7%-15% for stage I-II EC [29]. In a population-based cohort study including 1,630 women, Akesson et al. [30] reported an overall recurrence rate of 8.3% for EC stages FIGO IA to IIIC treated in accordance with current guidelines. ...
... High-risk EC often requires adjuvant radiation and systemic therapy [4,5]. Treatment options for advanced and recurrent ECs are limited, with radiotherapy and chemotherapy showing limited efficacy [6]. In this regard, EC molecular classification introduced by The Cancer Genome Atlas has enhanced diagnostic accuracy and refined risk stratification, identifying patients who can benefit from targeted therapies, ushering in a paradigm shift in patient treatment options [7,8]. ...
Article
Full-text available
HER2-targeted therapies have transformed the management of advanced or recurrent serous endometrial cancer (EC), leading to an increased clinical demand for HER2 testing. Despite its adoption in select academic centers, the global extent of such tumor testing is unclear. In this study, we report on the initial two-year experience of HER2 testing at a major academic center with a reference gynecologic oncology service and biomarker reference laboratory. All patients who underwent HER2 testing based on physician discretion, reflex HER2 testing, and reference laboratory requests were included. From February 2021 to October 2023, HER2 testing was performed on 192 tumor tissue samples from 180 EC patients. Serous carcinoma constituted 52% of samples, reflecting diagnostic challenges and limited therapeutic options for advanced EC. HER2 positivity was found in 28% of all cases and 30% of p53-aberrant cases. An immunohistochemistry (IHC) score of 3+ was found in 15% of samples, while IHC 2+ was found in 45% (13% IHC 2+/ISH+ and 32% IHC 2+/ISH-). The newly identified 'HER2-low' category comprised 46% of the samples. Heterogeneity was noted in 42% of HER2-positive cases, with complex patterns in 3%. NGS and HER2 IHC-FISH showed a 24% discordance, attributed to intratumoral heterogeneity, tumor cellularity, a small number of amplified cells, and the HER2/CEP17 ratio near the cut-off. This study offers real-world insights into HER2 testing in EC, highlighting the challenges and underscoring the need for standardized guidelines in specimen handling, proficiency testing, and scoring criteria to enhance patient management and therapeutic decision-making.
... Up to 20% of patients will develop local recurrence or metastatic disease [3][4][5][6][7]. Curative treatment options for metastatic EC are limited to women who only have locoregional metastases or isolated metastases [1,8,9]. For women with more advanced disease, only palliative options remain and are limited to the following: chemotherapy and hormonal therapy (HT) or novel therapies, including immunotherapy and other targeted treatment options [9][10][11]. ...
Article
Full-text available
Background: Response to hormonal therapy in advanced and recurrent endometrial cancer (EC) can be predicted by oestrogen and progesterone receptor immunohistochemical (ER/PR-IHC) expression, with response rates of 60% in PR-IHC > 50% cases. ER/PR-IHC can vary by tumour location and is frequently lost with tumour progression. Therefore, we explored the relationship between ER/PR-IHC expression and tumour location in EC. Methods: Pre-treatment tumour biopsies from 6 different sites of 80 cases treated with hormonal therapy were analysed for ER/PR-IHC expression and classified into categories 0-10%, 10-50%, and >50%. The ER pathway activity score (ERPAS) was determined based on mRNA levels of ER-related target genes, reflecting the actual activity of the ER receptor. Results: There was a trend towards lower PR-IHC (33% had PR > 50%) and ERPAS (27% had ERPAS > 15) in lymphogenic metastases compared to other locations (p = 0.074). Hematogenous and intra-abdominal metastases appeared to have high ER/PR-IHC and ERPAS (85% and 89% ER-IHC > 50%; 64% and 78% PR-IHC > 50%; 60% and 71% ERPAS > 15, not significant). Tumour grade and previous radiotherapy did not affect ER/PR-IHC or ERPAS. Conclusions: A trend towards lower PR-IHC and ERPAS was observed in lymphogenic sites. Verification in larger cohorts is needed to confirm these findings, which may have implications for the use of hormonal therapy in the future.
... На прогноз, тактику лечения пациенток, а также риск рецидива заболевания после первичного лечения влияют стадия и гистологические характеристики опухоли. При III и IV стадии 5-летняя выживаемость составляет 50 и 15 % соответственно [2,3]. Тактика лечения пациенток после прогрессирования РЭ зависит от некоторых факторов, однако в качестве 1-й линии системной терапии «золотым стандартом» себя зарекомендовала комбинация паклитаксела и карбоплатина [4]. ...
Article
Background. The inclusion of lenvatinib in the immunotherapy regimen for patients with MSS/pMMR endometrial cancer (EC) is due to its ability to modulate the tumor microenvironment, which allows the use of pembrolizumab in low-immunogenic tumors. However, only 30 % of patients with advanced or metastatic EC have a clinical response when treated with pembrolizumab and lenvatinib. In this regard, there is an obvious need to identify biomarkers that allow accurate selection of candidates for this type of therapy. Aim. To determine the predictive value of subpopulations of lymphocytes and macrophages, their expression of PD-1, expression of estrogen receptors, as well as vessel density in immunotargeted therapy for advanced or metastatic EC. Materials and methods. An open-label non-randomized observational association study was performed, involving a total of 22 patients with advanced or metastatic MSS/pMMR EC treated with pembrolizumab and lenvatinib. Duration of clinical effectiveness was used as a parameter to stratify patients. Using TSA-associated multiplex immunofluorescence, the proportions of CD8+ T lymphocytes, CD20+ B lymphocytes, FoxP3+ T regulatory lymphocytes and CD163+macrophages in tumor samples before the start of immunotargeted therapy were analyzed. Results. Three microenvironmental parameters were found to be associated with duration of clinical efficacy: the proportion of CD20+ B cells, the proportion of FoxP3+ T regulatory lymphocytes, and the ratio of CD8+/CD20+ lymphocytes in the tumor microenvironment. However, the CD8+/CD20+ lymphocyte ratio had the greatest predictive value; a value below 3.219 was associated with long clinical efficacy in patients with advanced or metastatic EC. Conclusion. The ratio of cytotoxic and B-lymphocytes in the microenvironment is a reliable predictor marker of the duration of the period of clinical effectiveness of immunotargeting therapy in advanced or metastatic EC.
... The decision to provide adjuvant treatment is based on the risk of the disease. Low-risk metastatic tumors can be treated with surgery alone, while in patients with high-risk metastatic tumors, additional adjuvant therapy is preferred [6]. ...
Article
Full-text available
Background Over the past decades, the rising incidence rates of endometrial cancer have made it a significant public health concern for women worldwide. Treatment strategies for endometrial cancer vary based on several factors such as stage, histology, the patient’s overall health, and preferences. However, limited amount of research on treatment patterns and potential correlations with sociodemographic characteristics among Hispanics is available. This study analyzes the treatment patterns for patients diagnosed with endometrial cancer in Puerto Rico. Methods A secondary database analysis was performed on endometrial cancer cases reported to the Puerto Rico Central Cancer Registry-Health Insurance Linkage Database from 2009 to 2015 (n = 2,488). The study population’s sociodemographic and clinical characteristics were described, along with an overview of the therapy options provided to patients receiving care on the island. Logistic regression models were used to evaluate the association of sociodemographic/clinical characteristics with treatment patterns stratified by risk of recurrence. Results In our cohort, most patients were insured through Medicaid and had a median age of 60 years. Almost 90% of patients received surgery as the first course of treatment. Surgery alone was the most common treatment for low-risk patients (80.2%). High-risk patients were more likely to receive surgery with radiotherapy and chemotherapy (24.4%). Patients with Medicare insurance were five times (HR: 4.84; 95% CI: 2.45–9.58; p < 0.001) more likely to receive surgery when compared with patients insured with Medicaid. In contrast, those with private insurance were twice as likely to receive surgery (HR: 2.38; 95% CI: 1.40–4.04; p = 0.001) when compared to those with Medicaid. Conclusion These findings provide insight into the treatment patterns for endometrial cancer in Puerto Rico and highlight the importance of considering factors such as disease risk when making treatment decisions. Addressing these gaps in treatment patterns can contribute to effective management of endometrial cancer.
... However, a delayed dia gnosis leads to progression of the tumor worsening the overall survival of the patient. Although relapse occurs in roughly 15% of cases, there is a lack of effective risk classification and limited progress in treating recurrent or metastatic disease [3]. This underscores the need to enhance the early-stage identification and stratification of patients with this form of cancer. ...
Article
Background: Endometrial carcinoma (EC) is the most common cancer of the female reproductive tract in developed countries. The prognosis and 5-year survival rates are closely tied to the stage diagnosis. Current routine diagnostic methods of EC are either lacking specificity or are uncomfortable, invasive and painful for the patient. As of now, the gold diagnostic standard is endometrial biopsy. Early and non-invasive diagnosis of EC requires the identification of new biomarkers of disease and a screening test applicable to routine laboratory diagnostics. The application of untargeted metabolomics combined with artificial intelligence and biostatistics tools has the potential to qualitatively and quantitatively represent the metabolome, but its introduction into routine diagnostics is currently unrealistic due to the financial, time and interpretation challenges. Fluorescence spectral analysis of body fluids utilizes autofluorescence of certain metabolites to define the composition of the metabolome under physiological conditions. Purpose: This review highlights the potential of fluorescence spectroscopy in the early detection of EC. Data obtained by three-dimensional fluorescence spectroscopy define the quantitative and qualitative composition of the complex fluorescent metabolome and are useful for identifying biochemical metabolic changes associated with endometrial carcinogenesis. Autofluorescence of biological fluids has the prospect of providing new molecular markers of EC. By integrating machine learning and artificial intelligence algorithms in the data analysis of the fluorescent metabolome, this technique has great potential to be implemented in routine laboratory diagnostics.
Article
Purpose: To investigate the cost-effectiveness of lenvatinib plus pembrolizumab (LP) compared to chemotherapy as a second-line treatment for advanced endometrial cancer (EC) from the United States and Chinese payers' perspective. Methods: In this economic evaluation, a partitioned survival model was constructed from the perspective of the United States and Chinese payers. The survival data were derived from the clinical trial (309-KEYNOTE-775), while costs and utility values were sourced from databases and published literature. Total costs, quality-adjusted life years (QALYs) and incremental cost-effectiveness ratio (ICER) were estimated. The robustness of the model was evaluated through sensitivity analyses, and price adjustment scenario analyses was also performed. Results: Base-case analysis indicated that LP wouldn't be cost-effective in the United States at the WTP threshold of $200 000, with improved effectiveness of 0.75 QALYs and an additional cost of $398596.81 (ICER $531392.20). While LP was cost-effective in China, with improved effectiveness of 0.75 QALYs and an increased overall cost of $62270.44 (ICER $83016.29). Sensitivity analyses revealed that the above results were stable. The scenario analyses results indicated that LP was cost-effective in the United States when the prices of lenvatinib and pembrolizumab were simultaneously reduced by 61.95% ($26.5361/mg for lenvatinib and $19.1532/mg for pembrolizumab). Conclusion: LP isn't cost-effective in the patients with advanced previously treated endometrial cancer in the United States, whereas it is cost-effective in China. The evidence-based pricing strategy provided by this study could benefit decision-makers in making optimal decisions and clinicians in general clinical practice. More evidence about budget impact and affordability for patients is needed.
Article
Full-text available
Endometriosis and adenomyosis behave similarly to cancer. No current treatments represent a cure, even if there are several options, including hormonal and surgical therapy. In advanced or recurrent pathologies, however, personalized treatment is necessary. We have found that due to the multiple common features, various therapeutic options have been used or studied for all three pathologies, with varying results. The objective of this review is to extract from the relevant literature the compounds that are used for endometriosis and adenomyosis characterized by malignant behavior, with some of these drugs being studied first in the treatment of endometrial cancer. Special attention is needed in the pathogenesis of these pathologies. Despite the multiple drugs that have been tested, only a few of them have been introduced into clinical practice. An unmet need is the cure of these diseases. Long-time treatment is necessary because symptoms persist, and surgery is often followed by postoperative recurrence. We emphasize the need for new, effective, long-term treatments based on pathogeny while considering their adverse effects.
Article
Full-text available
Objetivo: Comparar as taxas de recidiva local (cúpula vaginal) em pacientes portadoras de câncer de endométrio submetidas à histerectomia via convencional (aberta) versus histerectomia via laparoscópica. Métodos: Revisão narrativa da literatura a partir de estudos clínicos, de coorte e multicêntricos, publicados no período entre 2018 e 2022. Resultados: Nos estudos de coorte e multicêntricos retrospectivo revisados, as taxas de recidiva local foram semelhantes entre os grupos de mulheres com câncer de endométrio, operadas por via laparotômica e via laparoscópica, mostrando que a ocorrência não está associada à técnica cirúrgica escolhida para o tratamento. A eficácia e a segurança do procedimento laparoscópico estiveram associadas apenas às taxas de sobrevida e morbidade. Conclusão: Não existe diferenças nas taxas de recidiva local (cúpula vaginal) após tratamento de câncer de endométrio via convencional (laparotômica) e laparoscópica.
Article
Full-text available
Background Around 20% of women with endometrial cancer have advanced stage disease or suffer from a recurrence. For these women, prognosis is poor and palliative treatment options include hormonal therapy and chemotherapy. Lack of predictive biomarkers and suboptimal use of existing markers for response to hormonal therapy have resulted in overall limited efficacy. Objective To improve efficacy of hormonal therapy by relating immunohistochemical expression of estrogen (ER) and progesterone receptor (PR) and ER pathway activity scores to response to hormonal therapy. Study design Patients with advanced or recurrent endometrial cancer and available biopsies taken prior to start of hormonal therapy, were identified in 16 centers within the European Network for Individualized Treatment in Endometrial Cancer (ENITEC) and the Dutch Gynecologic Oncology Group (DGOG). Tumor tissue was analyzed for ER and PR expression and ER pathway activity using a qPCR-based mRNA model to measure the activity of ER-related target genes in tumor RNA. The primary endpoint was response rate defined as complete and partial response using RECIST criteria. Secondary endpoints were clinical benefit rate and progression free survival. Results Pretreatment biopsies with sufficient endometrial cancer tissue and complete response evaluation were available in 81 of 105 eligible cases. All patients with a response (n=22/81, 27.2%) had ER and PR expression>50%, resulting in a response rate of 32.3% (95%-CI: 20.9-43.7%) for ER-expresion>50% and 50.0% (95%-CI: 35.2-64.8%) for PR-expression>50%. Clinical benefit rate was 56.9% for ER-expression>50% (95%-CI: 44.9-68.9%) and 75.0% (95%-CI: 62.2-87.8%) for PR-expression>50%. Application of the ER pathway test to cases with PR-expression>50%, resulted in a response rate of 57.6% (95%-CI: 42.1-73.1%). After 2 years of follow-up, 34.3% (95%-CI: 20-48%) of cases with PR-expression >50% and 35.8% (95%-CI: 20-52%) of cases with ER pathway activity score >15 had not progressed. Conclusion Prediction of response to hormonal treatment in endometrial cancer improves substantially with a 50% cut-off level for PR-IHC and by applying a sequential test algorithm using PR-IHC and ER pathway activity scores. Results need to be validated in the PROMOTE-prospective study.
Article
Full-text available
Simple Summary Endometrial cancer (EC) represents 90% of uterine cancer and to date its standard clinical approach is still surgery and/or chemo- and radiotherapy. This mini-review illustrates the state of the art in the disease management. In particular, we aim to point out the following features: the hormonal nature of the pathology and the role of steroid receptors in EC promotion and progression; the importance of molecular and histopathological assessment for driving the clinic decision and the promising immunotherapeutic approaches with immune checkpoint blockade. Abstract EC is the most common cancer in the female genital tract in developed countries, and with its increasing incidence due to risk factors, such as aging and obesity, tends to become a public health issue. Although EC is a hormone-dependent neoplasm, there are no recommendations for the determination of steroid hormone receptors in the tumor tissue and no hormone therapy has ever been assessed in the adjuvant setting. Furthermore, its immune environment has been slightly characterized, but recent evidences point out how EC microenvironment may increase self-tolerance by reducing the recruitment of cytotoxic immune cells to the tumor site and/or modifying their phenotype, making these cells no longer able to suppress tumor growth. Here we highlight insights for EC management from diagnosis to a desirable trend of personalized treatment.
Article
Full-text available
Simple Summary This multicenter retrospective study aimed to describe the outcomes of patients with endometrial cancer after central pelvic/vaginal relapse treated with radical radiotherapy (RT). We included 139 patients with a median follow-up time of 6.66 years. Patients were treated with external beam radiotherapy to elective pelvic lymph-node regions and boost to the pelvic tumor. During follow-up, 55 (39.6%) patients developed a second relapse, the majority (75%) with disease sites outside the radiation field. Risk group at primary diagnosis and type of boost administration were independent predictors of progression-free and overall survival. Five-year overall survival for the whole cohort was 68% (95% CI: 59–75%). The majority of isolated pelvic recurrences in RT-naive women with EC can be successfully salvaged by RT but survival in high-risk patients remains suboptimal. Individualizing of adjuvant treatment in first line and better treatment alternatives at relapse are important to ultimately improve survival. Abstract (1) Background: This study evaluated the clinical outcome after salvage radiotherapy for first pelvic relapse after endometrial cancer (EC). (2) Methods: This multicenter retrospective study included EC patients with first central pelvic relapse without lymph node involvement treated with curative intent. Progression-free (PFS) and overall survival (OS) were calculated with the Kaplan–Meier method and possible predictive factors for risk of relapse and mortality were identified using the Cox model. (3) Results: We included 139 patients with median EQD2 (Equivalent Dose in 2 Gy fractions) to the clinical target volume of 70.0 Gy. During follow up of median 6.66 years, 39.6% patients developed a second relapse. Risk group classification at primary diagnosis based on histology, grading and FIGO stage and how the pelvic tumor boost was administered were independently associated with PFS and OS. Five-year OS was 68% (95% CI (59–75)) for the whole cohort. Five-year OS was 88% (95% CI (75–94)), 72% (95% CI (55–84)) and 38% (95% CI (15–60)) for the stage I low-, intermediate- and high-risk group, respectively. (4) Conclusions: The majority of central pelvic recurrences in RT-naive EC women can be successfully salvaged with radiotherapy. However, survival in patients with high-risk disease remains poor and warrants a more individualized approach to optimize outcome.
Article
Full-text available
A European consensus conference on endometrial carcinoma was held in 2014 to produce multi-disciplinary evidence-based guidelines on selected questions. Given the large body of literature on the management of endometrial carcinoma published since 2014, the European Society of Gynaecological Oncology (ESGO), the European SocieTy for Radiotherapy and Oncology (ESTRO), and the European Society of Pathology (ESP) jointly decided to update these evidence-based guidelines and to cover new topics in order to improve the quality of care for women with endometrial carcinoma across Europe and worldwide.
Article
Full-text available
Importance Deficient mismatch mutation repair mechanisms may sensitize endometrial cancers to anti–programmed death 1 (PD-1) therapies. Dostarlimab (TSR-042) is an investigational anti–PD-1 antibody that binds with high affinity to the PD-1 receptor. Objective To assess the antitumor activity and safety of dostarlimab for patients with deficient mismatch repair endometrial cancer. Design, Setting, and Participants This ongoing, open-label, single-group, multicenter study began part 1 on March 7, 2016, and began enrolling patients with deficient mismatch mutation repair endometrial cancer on May 8, 2017. Median follow-up was 11.2 months (range, 0.03 [ongoing] to 22.11 [ongoing] months; based on radiological assessments). Statistical analysis was performed July 8 to August 9, 2019. Interventions Patients received 500 mg of dostarlimab intravenously every 3 weeks for 4 doses, then 1000 mg every 6 weeks until disease progression, treatment discontinuation, or withdrawal. Main Outcomes and Measures The primary end point was objective response rate and duration of response by blinded independent central review using Response Evaluation Criteria in Solid Tumors, version 1.1. Results As of the data cutoff, 104 women (median age, 64.0 years [range, 38-80 years]) with deficient mismatch mutation repair endometrial cancers were enrolled and treated with dostarlimab. Of these, 71 had measurable disease at baseline and at 6 months or more of follow-up and were included in the analysis. There was a confirmed response in 30 patients (objective response rate, 42.3%; 95% CI, 30.6%-54.6%); 9 patients (12.7%) had a confirmed complete response, and 21 patients (29.6%) had a confirmed partial response. Responses were durable; the median duration of response was not reached (median follow-up was 11.2 months). The estimated likelihood of maintaining a response was 96.4% at 6 months and 76.8% at 12 months. Anemia (3 of 104 [2.9%]), colitis (2 of 104 [1.9%]), and diarrhea (2 of 104 [1.9%]) were the most common grade 3 or higher treatment-related adverse events. Conclusions and Relevance In this nonrandomized trial, dostarlimab was associated with clinically meaningful and durable antitumor activity with an acceptable safety profile for patients with deficient mismatch mutation repair endometrial cancers after prior platinum-based chemotherapy. Trial Registration ClinicalTrials.gov identifier: NCT02715284
Article
Full-text available
Purpose: MR-guided Radiation Therapy (MRgRT) allows for high-precision radiotherapy under real-time MR visualization. This enables margin reduction and subsequent dose escalation which may lead to higher tumor control and less toxicity. The Unity MR-linac (Elekta AB, Stockholm, Sweden) integrates a linear accelerator with a 1.5T diagnostic quality MRI and an online adaptive workflow. A prospective international registry was established to facilitate the evidence-based implementation of the Unity MR-linac into clinical practice, to systemically evaluate long-term outcomes, and to aid further technical development of MR-linac-based MRgRT. Methods and Results: In February 2019, the Multi-OutcoMe EvaluatioN of radiation Therapy Using the MR-linac study (MOMENTUM) started within the MR-linac Consortium. The MOMENTUM study is an international academic-industrial partnership between several hospitals and industry partner Elekta. All patients treated on the MR-linac are eligible for inclusion in MOMENTUM. For participants, we collect clinical patient data (e.g., patient, tumor, and treatment characteristics) and technical patient data which is defined as information generated on the MR-linac during treatment. The data are captured, pseudonymized, and stored in an international registry at set time intervals up to two years after treatment. Patients can choose to provide patient-reported outcomes and consent to additional MRI scans acquired on the MR-linac. This registry will serve as a data platform that supports multicenter research investigating the MR-linac. Rules and regulations on data sharing, data access, and intellectual property rights are summarized in an academic-industrial collaboration agreement. Data access rules ensure secure data handling and research integrity for investigators and institutions. Separate data access rules exist for academic and industry partners. This study is registered at ClinicalTrials.gov with ID: NCT04075305 (https://clinicaltrials.gov/ct2/show/NCT04075305). Conclusion: The multi-institutional MOMENTUM study has been set up to collect clinical and technical patient data to advance technical development, and facilitate evidenced-based implementation of MR-linac technology with the ultimate purpose to improve tumor control, survival, and quality of life of patients with cancer.
Article
Full-text available
Oestrogen receptor (ER) expression is a prognostic biomarker in endometrial cancer (EC). However, expression does not provide information about the functional activity of the ER pathway. We evaluated a model to quantify ER pathway activity in EC, and determined the prognostic relevance of ER pathway activity. ER pathway activity was measured in two publicly available datasets with endometrial and EC tissue, and one clinical cohort with 107 samples from proliferative and hyperplastic endometrium and endometrioid-type EC (EEC) and uterine serous cancer (USC). ER pathway activity scores were inferred from ER target gene mRNA levels from Affymetrix microarray data (public datasets), or measured by qPCR on formalin-fixed paraffin-embedded samples (clinical cohort) and related to ER expression and outcome. ER pathway activity scores differed significantly throughout the menstrual cycle supporting the validity of the pathway test. The highest ER pathway scores were found in proliferative and hyperplastic endometrium and stage I EEC, whereas stage II–IV EEC and USCs had significantly lower levels. Low ER pathway activity was associated with recurrent disease, and added prognostic value in patients with low ER expression. The ER pathway test reflects activity of the ER pathway, and may improve prediction of outcome in EC patients.
Article
Full-text available
PURPOSE The oligometastatic paradigm hypothesizes that patients with a limited number of metastases may achieve long-term disease control, or even cure, if all sites of disease can be ablated. However, long-term randomized data that test this paradigm are lacking. METHODS We enrolled patients with a controlled primary malignancy and 1-5 metastatic lesions, with all metastases amenable to stereotactic ablative radiotherapy (SABR). We stratified by the number of metastases (1-3 v 4-5) and randomized in a 1:2 ratio between palliative standard-of-care (SOC) treatments (arm 1) and SOC plus SABR (arm 2). We used a randomized phase II screening design with a primary end point of overall survival (OS), using an α of .20 (wherein P < .20 indicates a positive trial). Secondary end points included progression-free survival (PFS), toxicity, and quality of life (QOL). Herein, we present long-term outcomes from the trial. RESULTS Between 2012 and 2016, 99 patients were randomly assigned at 10 centers internationally. The most common primary tumor types were breast (n = 18), lung (n = 18), colorectal (n = 18), and prostate (n = 16). Median follow-up was 51 months. The 5-year OS rate was 17.7% in arm 1 (95% CI, 6% to 34%) versus 42.3% in arm 2 (95% CI, 28% to 56%; stratified log-rank P = .006). The 5-year PFS rate was not reached in arm 1 (3.2%; 95% CI, 0% to 14% at 4 years with last patient censored) and 17.3% in arm 2 (95% CI, 8% to 30%; P = .001). There were no new grade 2-5 adverse events and no differences in QOL between arms. CONCLUSION With extended follow-up, the impact of SABR on OS was larger in magnitude than in the initial analysis and durable over time. There were no new safety signals, and SABR had no detrimental impact on QOL.
Article
Background there is no consensus on the cut-off for positivity of estrogen receptor (ER) and progesterone receptor (PR) in endometrial cancer (EC). Therefore we determined the cut-off value for ER and PR with the strongest prognostic impact on outcome. Methods immunohistochemical expression of ER and PR was scored as a percentage of positive EC cell nuclei. Cut-off values were related to disease-specific (DSS) and disease-free survival (DFS) using sensitivity, specificity and multivariable regression analysis. The results were validated in an independent cohort. Results the study cohort (n=527) included 82% grade 1-2 and 18% grade 3 ECs. Specificity for DSS and DFS was highest for the cut-off values 1-30%. Sensitivity was highest for the cut-offs 80-90%. ER and PR expression were independent markers for DSS at cut-off values of 10% and 80%. Consequently, three subgroups with distinct clinical outcome were identified: ER/PR 0-10%: unfavorable outcome (5-year-DSS 75.9-83.3%); ER/PR 20-80%: intermediate outcome (5-year-DSS 93.0-93.9%) and ER/PR 90-100%: favorable outcome (5-year-DSS 97.8-100%). The association between ER/PR subgroups and outcome was confirmed in the validation cohort (n=265). Conclusions we propose classification of ER and PR expression according to a high risk (0-10%), intermediate risk (20-80%) and low risk (90-100%) group.
Article
PURPOSE Limitations of the paclitaxel-doxorubicin-cisplatin (TAP) regimen in the treatment of endometrial cancer include tolerability and cumbersome scheduling. The Gynecologic Oncology Group studied carboplatin plus paclitaxel (TC) as a noninferior alternative to TAP. METHODS GOG0209 was a phase III, randomized, noninferiority, open-label trial. Inclusion criteria were stage III, stage IV, and recurrent endometrial cancers; performance status 0-2; and adequate renal, hepatic, and marrow function. Prior radiotherapy and/or hormonal therapy were permitted, but chemotherapy, including radiosensitization, was not. Patients were treated with doxorubicin 45 mg/m ² and cisplatin 50 mg/m ² (day 1), followed by paclitaxel 160 mg/m ² (day 2) with granulocyte colony-stimulating factor or paclitaxel 175 mg/m ² and carboplatin area under the curve 6 (day 1) every 21 days for seven cycles. The primary endpoint was overall survival (OS; modified intention to treat). Progression-free survival (PFS), health-related quality of life (HRQoL), and toxicity were secondary endpoints. RESULTS From 2003 to 2009, 1,381 women were enrolled. Noninferiority of TC to TAP was concluded for OS (median, 37 v 41 months, respectively; hazard ratio [HR], 1.002; 90% CI, 0.9 to 1.12), and PFS (median, 13 v 14 months; HR, 1.032; 90% CI, 0.93 to 1.15). Neutropenic fever was reported in 7% of patients receiving TAP and 6% of those receiving TC. Grade > 2 sensory neuropathy was recorded in 26% of patients receiving TAP and 20% receiving TC ( P = .40). More grade ≥ 3 thrombocytopenia (23% v 12%), vomiting (7% v 4%), diarrhea (6% v 2%), and metabolic (14% v 8%) toxicities were reported with TAP. Neutropenia (52% v 80%) was more common with TC. Small HRQoL differences favored TC. CONCLUSION With demonstrated noninferiority to TAP, TC is the global first-line standard for advanced endometrial cancer.