Next Article in Journal
Recent Advances in Therapeutic Strategies to Improve Colorectal Cancer Treatment
Previous Article in Journal
Comparison of Immunotherapy versus Targeted Therapy Effectiveness in BRAF-Mutant Melanoma Patients and Use of cGAS Expression and Aneuploidy as Potential Prognostic Biomarkers
Previous Article in Special Issue
Impact of Patient Body Mass Index on Post-Operative Recovery from Robotic-Assisted Hysterectomy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Advances in Endometrial Cancer Prevention, Early Diagnosis and Treatment

by
Holly Baker-Rand
1 and
Sarah J. Kitson
1,2,*
1
Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
2
Manchester Academic Health Science Centre, Department of Obstetrics and Gynaecology, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK
*
Author to whom correspondence should be addressed.
Cancers 2024, 16(5), 1028; https://doi.org/10.3390/cancers16051028
Submission received: 15 February 2024 / Revised: 26 February 2024 / Accepted: 27 February 2024 / Published: 1 March 2024

Abstract

:

Simple Summary

Endometrial cancer is the fourth most common female malignancy in high socioeconomic index nations and the sixth most common cancer in women worldwide. The disease incidence has increased globally by 132% in the last 30 years, and this trend is set to continue in light of an ageing population and increasing levels of obesity and diabetes. Although more women than ever before are dying of endometrial cancer, the mortality rates are falling due to recent advances in early diagnosis and treatment. Our understanding of the molecular drivers of endometrial cancer has increased substantially, and doctors are now, for the first time, starting to translate this knowledge into the true personalisation of care. This review summarises the recent advances in the field and identifies avenues for future exploration.

Abstract

Endometrial cancer is the sixth commonest cancer in women worldwide, with over 417,000 diagnoses in 2020. The disease incidence has increased by 132% over the last 30 years and is set to continue to rise in response to an ageing population and increasing global rates of obesity and diabetes. A greater understanding of the mechanisms driving endometrial carcinogenesis has led to the identification of potential strategies for primary disease prevention, although prospective evaluation of their efficacy within clinical trials is still awaited. The early diagnosis of endometrial cancer is associated with improved survival, but has historically relied on invasive endometrial sampling. New, minimally invasive tests using protein and DNA biomarkers and cytology have the potential to transform diagnostic pathways and to allow for the surveillance of high-risk populations. The molecular classification of endometrial cancers has been shown to not only have a prognostic impact, but also to have therapeutic value and is increasingly used to guide adjuvant treatment decisions. Advanced and recurrent disease management has also been revolutionised by increasing the use of debulking surgery and targeted treatments, particularly immunotherapy. This review summarises the recent advances in the prevention, diagnosis and treatment of endometrial cancer and seeks to identify areas for future research.

1. Introduction

The endometrial cancer landscape has changed markedly over the last 30 years, with a notable rise in disease incidence and increasing patient complexity. In 2020, there were over 417,000 new cases of endometrial cancer diagnosed worldwide and 97,370 disease-related deaths, representing a substantial health burden [1]. These numbers are set to increase further in light of an ageing global population and rising rates of obesity and diabetes [2]. A better understanding of the mechanisms through which these risk factors drive endometrial carcinogenesis has provided an opportunity to develop targeted interventions for primary disease prevention, with the aim of halting this increase. Although the number of endometrial cancer deaths has nearly doubled since 1990, the age-standardised mortality rate has significantly decreased in almost all global regions, reflecting advances in early diagnosis and treatment [3]. Whilst the majority of women are diagnosed with early-stage disease and can expect to be cured of their endometrial cancer, up to 20% of women continue to have extra-uterine disease at presentation, with only 15% of women with stage IV disease alive five years after diagnosis [2]. Early diagnosis, therefore, remains imperative, and non-invasive tests designed to exclude endometrial cancer in the majority of women with postmenopausal bleeding are rapidly increasing in number and are likely to be more acceptable to women than endometrial biopsies. Of concern is the widening disparity in survival between White, Black, Asian and Hispanic women, which may be related as much to the lack of access to endometrial cancer treatments as the differences in tumour biology [4,5]. A greater understanding of the genetic changes driving endometrial carcinogenesis will hopefully be used to address this gap, but this has already begun to be used to personalise treatments and to identify those women with Lynch syndrome, for whom their endometrial cancer represents a sentinel malignancy [6]. This review summarises these recent advances in endometrial cancer prevention, diagnosis and treatment and seeks to identify the outstanding research questions to be addressed in the next 30 years.

2. Advances in Endometrial Cancer Prevention

Given the strong association with modifiable risk factors, endometrial cancer appears eminently suited to primary disease prevention, with modelling suggesting that up to 60% of endometrial cancer cases could be potentially prevented [7,8,9]. An increased understanding of the mechanisms driving endometrial carcinogenesis, namely unopposed oestrogen, insulin resistance and chronic inflammation, has led to the proposal of a number of interventions designed to reduce endometrial cancer incidence, albeit with data on their efficacy largely limited to retrospective observational studies [10,11].

2.1. Weight Management

Obesity has the strongest link with endometrial cancer of the twenty most common tumour types and is plausibly implicated in 34% of diagnoses [7]. Conversely, weight loss, by reducing adiposity and the aromatase-induced conversion of androgens into oestrogen, improving insulin sensitivity and lowering the levels of inflammation, is associated with a reduction in the endometrial cancer risk [11]. Observational data from the Women’s Health Initiative suggests that intentional weight loss of as little as 5% bodyweight over a three year period is associated with a 39% (95%CI 12–58%) reduction in endometrial cancer incidence [12]. Interestingly, whilst women with obesity had the greatest benefit (HR 0.44, 95%CI 0.25–0.78), weight loss was also beneficial for women with a BMI within the normal range (HR 0.61, 95%CI 0.27–1.38). Achieving weight loss through lifestyle modification is feasible, but challenging to maintain and risks weight cycling, which appears to be more detrimental to endometrial cancer risk than a stable higher weight [13]. Certainly, the seven hours of jogging a week required to reduce endometrial cancer risk by 10% is not likely to be achievable for the majority of women [14].
The focus has, therefore, shifted to the new anti-obesity medications (AOMs) that have been developed in recent years and which appear to lead to more significant and sustained weight loss in comparison with lifestyle interventions and older AOMs such as orlistat [15,16]. Glucagon-like peptide 1 (GLP-1) agonists, such as semaglutide and liraglutide, improve insulin sensitivity, delay gastric emptying and decrease a person’s appetite [17]. The mean weight change at two years was −15.2% with weekly semaglutide compared with −2.6% with a placebo in a STEP 5 randomised controlled trial (RCT), when used in combination with a behavioural intervention (p < 0.0001) [18]. Liraglutide appears to be similarly efficacious, with 51.8% of participants achieving ≥5% bodyweight loss compared with 24% of those treated with a placebo in an SCALE RCT (p < 0.0001) [19]. The side effects include gastrointestinal disruption, skin reactions, atrioventricular blockage, and rarely, pancreatitis, and long-term tolerance and safety have yet to be established. Whether the effects of GLP-1 agonists on bodyweight and insulin sensitivity can also translate into a reduction in endometrial cancer risk remains to be seen and should be included as a secondary outcome measure in long-term cohort studies.
Bariatric surgery alters the anatomy of the digestive system to restrict the capacity of the stomach, reduce nutrient absorption and induce early satiety [20]. It is the most effective intervention for obesity identified to date, leading to the long-term weight loss of up to 29 kg depending upon the exact procedure performed [21]. Wilson et al. demonstrated in their meta-analysis that bariatric surgery is effective in reducing the risk of subsequent endometrial cancer by 62% (pooled relative risk 0.38, 95%CI 0.26–0.55) [22]. Despite this, endometrial cancer prevention is not currently an indication for bariatric surgery, potentially because of the risk of long-term complications, including malabsorption, nutritional deficiencies, small bowel obstruction, dumping syndrome and gastric or stomal stenosis and resource issues limiting its availability [23].

2.2. Hormonal Chemoprevention

The beneficial effects of oral contraceptives on endometrial cancer risk have been known for the last 20 years, with every five years of use associated with a 24% reduction in the disease risk [24]. Importantly, this effect appears to persist for up to 30 years after the discontinuation of use. Whilst oestrogen-containing preparations are not advisable for women with a BMI > 35 kg/m2 due to an elevated risk of arterial and venous thrombotic events, the combined oral contraceptive pill is recommended by the international consensus group for women with Lynch syndrome requiring contraception due to its beneficial effects on both the endometrial and ovarian cancer risks [25,26]. Progestin-only contraception is also likely to be beneficial, although the discontinuation rates are often higher due to irregular bleeding [27]. The levonorgestrel-releasing intra-uterine system (LNG-IUS) may well be the most effective endometrial cancer prevention measure, with large-scale observational studies describing up to a 78% reduction in endometrial cancer risk among the users, particularly if used long-term [28,29]. It certainly appears to reduce endometrial proliferation, even in women with a BMI ≥ 40 kg/m2, with modelling suggesting that it could be a cost-effective approach for primary disease prevention in high-risk women [30,31]. Reassuringly, the previously raised concerns about an increased risk of postmenopausal breast cancer with LNG-IUS use were not confirmed in a recent meta-analysis [32]. Adequately powered clinical trials are now required to determine whether the LNG-IUS is both effective at reducing the incidence of endometrial cancer and is sufficiently acceptable to women for it to be used in routine practice.

2.3. Aspirin

Aspirin, a cycloxyengase-2 inhibitor, has anti-inflammatory effects and acts to reduce the aromatase and oestrogen levels and increase apoptosis [33,34]. Whilst women within the general population may benefit from only a small reduction in endometrial cancer risk from long-term aspirin use (8–11%), this may prove to be a more effective strategy in women with obesity (relative risk reduction 20–44%) [35]. The CAPP2 study demonstrated a clear reduction in the colorectal cancer risk with regular aspirin use in individuals with Lynch syndrome, but this RCT was insufficiently powered to assess the benefit to women with regard to the endometrial cancer risk [36]. The results were, however, encouraging (hazard ratio 0.50, 95%CI 0.22–1.11) and should be investigated further.

2.4. Metformin

Much interest has been expressed in the re-purposing of metformin, an oral biguanide and insulin-sensitiser, in the management of endometrial cancer. Despite early promising results in single-arm studies, metformin did not appear to reduce endometrial proliferation when evaluated within a more methodologically robust RCT [37]. These findings have been confirmed in a Cochrane review, in which it was noted that there was insufficient evidence to support the use of metformin either alone or in combination with progestin therapy for the management of endometrial hyperplasia [38]. The authors did note, however, that only two trials totally 59 patients were eligible for inclusion in their review, making it difficult to draw generalisable conclusions. A meta-analysis of six studies also found that metformin use was not associated with a reduction in the endometrial cancer risk (odds ratio 1.05, 95%CI 0.82–1.35), even when adjusting for the confounding variable of diabetes [39]. A recently published feMMe trial again demonstrated the limited cytostatic effect of metformin on the endometrium, with no increase in the effectiveness of the LNG-IUS for the management of early-stage endometrial cancer with the addition of metformin [40]. Together, these results should dissuade researchers from pursuing metformin for the chemoprevention or treatment of endometrial cancer.

2.5. Identifying High-Risk Women

Identifying high-risk individuals for targeted endometrial cancer prevention is imperative in order to maximise the benefits and minimise the risk of harm from long-term intervention. Women with Lynch syndrome represent the highest-risk group, with a 40–60% lifetime risk of endometrial cancer depending upon the underlying genetic variant [41]. The widespread adoption of reflex immunohistochemistry for mismatch repair genes (MLH1, MSH2, MSH6 and PMS2) on endometrial cancers prompts the identification of affected individuals, who will, themselves, benefit from surveillance for colorectal malignancies and whose affected female relatives should be offered a risk-reducing hysterectomy from the age of 40 years [26].
Only 3% of endometrial cancers are, however, related to Lynch syndrome, with a much greater number of women within the general population at moderate–high risk of the disease secondary to polygenic and environmental factors. Offering an intervention to this group has the potential to dramatically reduce the endometrial cancer incidence. To date, four risk prediction models have been developed with the aim of stratifying the endometrial cancer risk within the general population, of which three models have been externally validated [42,43,44,45]. All are based on similar epidemiological risk factors, with or without the addition of serum biomarkers, with the E2C2 and PRECISION models demonstrating the highest discrimination (0.64–0.69) and best calibration (E/0 1.03–1.09) via external validation [44,45]. The advantage of the PRECISION model is its generalisability to non-White ethnic groups and suitability for use in both pre- and postmenopausal women, although, currently, it has only been validated within a UK population. In this setting, it outperformed the other published models in decision curve analysis and was associated with a greater net benefit than offering endometrial cancer prevention to all the women. An endometrial cancer risk assessment appeals to women who are keen to know their own individualised risk, in theory, and are willing to make lifestyle changes or have an LNG-IUS inserted if it will be of benefit [46]. The uptake of interventions to reduce the endometrial cancer risk need, therefore, to be urgently assessed within the context of a clinical trial.

3. Advances in Early Diagnosis

The majority of cases of endometrial cancer are diagnosed among postmenopausal women who frequently present with postmenopausal bleeding [10]. Transvaginal ultrasound in combination with endometrial sampling enables the histological diagnosis of endometrial cancer and can accurately exclude the presence of disease. Endometrial biopsies are, however, invasive and have a failure rate of around 11% due to inadequate samples and cervical stenosis [47]. Innovations in diagnostic tests are underway to identify alternative methods of identifying high-risk women for further testing and which can be used to reassure those at low risk of the disease. Peripheral blood, cervicovaginal fluid and urine offer potential sources of DNA or protein biomarkers and cells for cytological assessment.

3.1. Peripheral Blood

Human epididymis protein 4 (HE4) has emerged as the most promising of a number of serum diagnostic endometrial cancer biomarkers examined to date. Two meta-analyses have demonstrated a relatively modest pooled sensitivity of 65%, but with a higher specificity at 91%, albeit with marked inter-study heterogeneity [48,49]. The addition of further biomarkers, including cancer antigen 125 (CA125), and anthropometric data, such as BMI, has not been shown to improve the test performance [50]. These results suggest that the HE4 levels alone are unlikely to be sufficiently informative for them to be used in a diagnostic test, but that they may have a role as a triage tool for high-risk populations.
There has been increasing interest in the role of plasma cell free DNA (cfDNA), circulating tumour DNA (ctDNA) and circulating microRNA (miRNA) for the detection of endometrial cancer. The next-generation sequencing of cfDNA using a targeted four-gene panel (CTNNB1, K-ras, PTEN and PIK3CA) identified mutations within the plasma that matched those within the corresponding endometrial tumour in 33% of women, although this could only detect 18% of mutations in women with early-stage disease [51]. A recent study using ctDNA detected the hypermethylation of zinc finger and SCAN domain containing 12 (ZSCAN12) and/or oxytocin (OXT) in 9/11 and 5/20 women with advanced and non-advanced endometrial cancer, respectively, giving a sensitivity of 98%, specificity of 97% and an area under the curve (AUC) of 0.99 [52]. Fan et al. assessed the miRNA signatures in 92 endometrial cancer subjects and 102 control subjects and externally validated the identified signatures in three large datasets. Six miRNAs (miR-143-3p, miR-195-5p, miR-20b-5p, miR-204-5p, miR-423-3p and miR-484) were overexpressed in endometrial cancer, with an AUC via external validation of 0.97 [53]. The observed discrepancy in test performance based on the stage of disease is not surprising given that localised disease is unlikely to shed sufficient ctDNA into the blood for it to be reliably detected. These biomarkers may, therefore, be better suited to identifying aggressive and potentially recurrent endometrial cancer rather than being used for the early diagnosis of disease.
Advances in the use of high-throughput technology have led to a rapid increase in the number of registered studies aiming to identify novel genomic, transcriptomic, proteomic and metabolomic blood biomarkers to be used in the early diagnosis of endometrial cancer. Single biomarkers may not be sufficiently informative to be used in clinical practice, and many of the ‘omic signatures’ identified have yet to be externally validated. This remains, however, an active area of research.

3.2. Uterine and Cervicovaginal Fluid

Uterine lavage fluid has been shown to have good sensitivity and specificity for endometrial cancer detection; however, it needs to be collected via hysteroscopy and is associated with significant discomfort for women [54]. Cervicovaginal samples are easier to obtain and can be as informative for the detection of an endometrial cancer as uterine lavage due to the shedding of malignant cells into the lower genital tract. The PapSEEK test, which incorporates assays for mutations in 18 genes as well as aneuploidy, has been shown to identify 81% of endometrial cancers when conducted during a routine Pap test, including 78% with early-stage disease [55]. The sensitivity increased to 93%, with a specificity of 100% when a Tao brush was used to obtain the samples. The Tao brush needs to be inserted up to the uterine fundus though and is associated with discomfort and a high failure rate. Alternatively, vaginal tampons can be used to collect cervicovaginal fluid, representing an inexpensive, easy-to-use self-collection system that is more likely to appeal to women. The samples can be analysed by next-generation sequencing to identify somatic mutations [56] or, of notable promise, undergo methylation testing [57]. The WID-qEC test, a three-marker test that appraises DNA methylation in the gene regions of GYPC and ZSCAN12 has been shown to outperform the transvaginal ultrasound measurement of endometrial thickness in the diagnosis of endometrial cancer (AUC WID-qEC 0.94 vs. ET measurement 0.87) among women with abnormal uterine bleeding [57]. The high negative predictive value of the test means that it could be used to reduce the number of women requiring invasive histological assessments.

3.3. Urine and Vaginal Cytology

Intact cells shed from the upper genital tract can also be detected in the vaginal fluid and, interestingly, within urine. This is thought to be related to the contamination of voided urine samples during self-collection, with higher cellular loads potentially more likely to be found in the samples collected from women with concurrent abnormal uterine or postmenopausal bleeding [58]. Cytological assessment is labour-intense and requires specialist interpretation, but has been shown within a diagnostic accuracy study to have a combined sensitivity of 91.7% (95%CI 85.0–96.1%) and specificity of 88.8% (95%CI 81.2–94.1%) for the detection of gynaecological malignancies, the majority of which were endometrial cancers [58]. The high negative predictive value (91.4%, 95%CI 84.9–95.2%) of urine and vaginal cytology may mean that it can be used to identify women for future investigations, reducing the number of women with abnormal uterine bleeding who require an invasive endometrial biopsy. There is clear potential for the analysis of these samples to extend to adjunct immunocytochemistry and genomic analysis, with the role of artificial intelligence to support this currently under investigation.
All of these novel diagnostic techniques have the potential to change the landscape of endometrial cancer diagnosis. Minimally invasive tests are likely to reduce pain and distress for women and could be used to reassure women at low risk of the disease, meaning that only those most likely to have an underlying endometrial cancer are required to undergo endometrial biopsy. These tests could also be used repeatedly to screen for endometrial cancer in women at high risk of the disease. At present, however, these early diagnostic techniques are only being used within the context of research studies, with evidence of their clinical utility available from a limited number of small cohorts of women who have already been diagnosed with endometrial cancer or presenting with postmenopausal bleeding. Large prospective multicentre trials are urgently needed to confirm the diagnostic accuracy of these techniques if they are to be rolled out into routine clinical practice.

4. Surgery

Sentinel lymph node mapping has transformed surgery for endometrial cancer since it was first described by Burke et al. in 1996 [59]. Whilst the first pilot study involved the use of isosulfan blue dye injected into the subserosal myometrium, the more recent studies have used a variety of tracers alone or in combination (blue dye, indocyanine green (ICG) and technetium-99m) and a number of different routes of injection (subserosal, cervical and endometrial via hysteroscopy). A Cochrane review failed to demonstrate the significant effect of type or route of dye injection on the number of sentinel lymph nodes identified, although the authors noted that the number of studies using the same protocol that could be combined in a meta-regression analysis was small [60]. Accessing the cervix for injection is certainly easier than pursuing peri-tumoural injection, and ICG may offer an improved sentinel lymph node detection rate over that of blue dye [61]. This certainly has the added advantage of compatibility with robotic platforms now in widespread use. Given the high detection rate (mean 86.9%, 95%CI 82.9–90.8%) and sensitivity (pooled 91.8%, 95%CI 86.5–95.1%) of sentinel lymph node detection secondary to ultrastaging combined with a low false-positive rate and reduced risk of lymphoedema, many international societies have recommended sentinel lymph node biopsy replace pelvic node dissection as part of endometrial cancer staging [60,62,63]. Whilst a therapeutic benefit from pelvic lymphadenectomy in women with presumed early-stage endometrial cancer has never been demonstrated, it does allow for adjuvant treatment to be used selectively among those at the greatest risk of disease recurrence [64].
Robotic surgery is now much more widely available and is being used increasingly to treat women with endometrial cancer. Despite the concerns about cost and longer operating times, it does appear to offer a number of advantages over conventional (straight-stick) laparoscopy, particularly in women with a BMI ≥ 40 kg/m2. In particular, its use is associated with a shorter hospital stay and lower rate of conversion to laparotomy (robotic surgery pooled conversion rate 3.8%, 95%CI 1.4–9.9 vs. laparoscopic surgery 7.0%, 95%CI 3.2–14.5), potentially because of the use of lower intra-abdominal pressure and consequent reduced intolerance of the Trendelenburg position [65,66]. Importantly, similar oncological outcomes have been demonstrated for patients undergoing robotic, laparoscopic and open surgery for early-stage endometrial cancer in retrospective series, although the data from prospective longitudinal studies are still awaited [66,67]. There are also benefits for surgeons, with improved ergonomics and reduced muscle usage, especially when operating on patients with a high BMI [68].
Whilst surgery remains the primary treatment modality for women diagnosed with early-stage endometrial cancer, the role of cytoreductive surgery in advanced disease continues to be contentious. Barlin et al. demonstrated in their meta-analysis of 14 retrospective cohort studies that a 10% increase in the complete cytoreduction rate for women with advanced endometrial cancer was associated with a 9.3-month increase in overall survival [69]. Whether the presence of any residual disease at the end of surgery negatively impacts upon survival remains under debate, largely because of the widely varying definitions used to describe optimal cytoreduction in the literature [69,70]. Over the last 15 years there has been an increase in the use of neoadjuvant chemotherapy in the treatment of advanced endometrial cancer, with the hope of trying to improve on the otherwise disappointing rates of maximal cytoreduction with primary surgery [70]. There are, however, only limited data demonstrating a reduction in surgical morbidity and operating time and a modest improvement in rates of optimal cytoreduction with this approach [71]. This is perhaps not surprising given the diverse patients and surgical complexity being studied. Evidence on the long-term oncological outcomes of women undergoing neoadjuvant chemotherapy and interval surgery for advanced endometrial cancer should be a research priority.

5. Adjuvant Treatment

Despite the lack of prospective clinical trial data on the use of molecular profiling to determine management, the FIGO have elected to incorporate the four endometrial cancer molecular subgroups (POLEmut, MMRd, NSMP and p53abn) into their most recent staging [72,73]. The implementation of the new system has the potential to lead to profound changes in the adjuvant treatment of endometrial cancer, but we should await the results of an ongoing multi-arm RAINBO trial [74]. This umbrella study consists of three randomised controlled trials and one prospective two arm phase II trial and will compare radiotherapy and chemotherapy with and without the maintenance of Olaparib (p53abn), radiotherapy alone or in combination with durvalumab (MMRd), radio- and chemotherapy with radiotherapy and progesterone maintenance (NSMP) and radiotherapy with no additional treatment (POLEmut) in an adjuvant setting. Survival and quality of life outcomes at three and five years will be assessed.
In the interim, the molecular characterisation of endometrial cancers is already being encouraged to be considered when making decisions about adjuvant treatment and provides important prognostic data (Table 1) [62]. The low rate of recurrence seen with POLEmut tumours suggests that an adjuvant treatment may not be required for patients with early-stage disease and a pathogenic POLE mutation [75]. On the other hand, the escalation of adjuvant treatment to include combined chemotherapy and radiotherapy appears to be beneficial for women with stage III disease and p53abn tumours, and this is associated with 11.3% and 22.4% increases in the 5-year failure-free survival rate, respectively, in the PORTEC-3 trial [76,77]. The absence of data to support a benefit of adjuvant chemotherapy for women with MMRd tumours, even in the presence of advanced disease, is increasingly being used to justify the avoidance of the systematic toxicity associated with its use [77].

6. Immunotherapy

After years of limited treatment options for advanced and recurrent endometrial cancer, the widespread approval of immunotherapy by international drug agencies (the FDA, EMA and NICE) has been heralded as a major advance for patients. Both pembrolizumab and dostarlimab bind to the PD-1 receptor and block the activation of the PDL-1 and PDL-2 pathways, which are critical in controlling immune tolerance within the tumour microenvironment. Microsatellite high (MSI-H) and MMRd tumours demonstrate the upregulation of PD-L1 and, therefore, increased sensitivity to immune checkpoint inhibition [78]. This was confirmed within the Keynote-158 study, in which a 48% objective response rate to pembrolizumab was observed in 79 patients with MSI-H/MMRd endometrial cancers, a significant proportion of whom was heavily pre-treated [79]. A similar response rate was observed with dostarlimab in this patient group (42.3%) in the GARNET trial, with an apparently sustained response [80]. Whilst both drugs are, therefore, licensed for use by women with advanced or recurrent MSI-H/MMRd endometrial cancer (Table 2) who have progressed on or following platinum-based chemotherapy, the cost may well direct drug choice, and tolerability is a concern [81,82]. Only dostarlimab has been approved for use alongside chemotherapy in the primary treatment of advanced MSI-H/MMRd endometrial cancer, following the RUBY trial, which demonstrated an improvement in progression-free survival at 24 months from 15.7% (95%CI 7.2–27.0) to 61.4% (95%CI 46.3–73.4%) with the addition of the drug [83]. The combination of pembrolizumab with Lenvatinib (a tyrosine kinase inhibitor) appears to be equally efficacious in women with MMR-proficient endometrial cancers as it does with those with MMRd tumours, resulting in a median 3.4 month prolongation of progression-free survival compared with chemotherapy alone [82]. Identifying the predictive biomarkers for this regime is now under investigation [84].

7. Medical Management

Increasing numbers of women are opting to undergo the medical management of their endometrial cancer, either as a means of preserving fertility or because of significant peri-operative risks driven by obesity and associated co-morbidities. Intrauterine progestin has a similar response rate to oral progesterones, but may be associated with fewer side effects, and hence, improved compliance [85,86]. Women with atypical hyperplasia are more likely to achieve a complete response than those with endometrial cancer, and molecular classification may well be used in the future to guide therapy [87,88]. Simultaneous weight loss either through dieting or bariatric surgery appears to improve the effectiveness of progesterone therapy, with women who lost > 10% bodyweight being four times more likely to respond than those who lost ≤ 10% bodyweight [89]. This may also have a beneficial effect on improving fertility, as well as reducing the risks associated with hysterectomy, particularly if there is disease relapse.

8. The Future of Endometrial Cancer Research

Significant advances in our understanding of endometrial carcinogenesis and the changing demographics of those diagnosed with the disease have driven seismic shifts in the way endometrial cancer is diagnosed and treated and has identified, for the first time, the ways in which this disease could be prevented. The increasing personalisation of care based on patients’ and tumour characteristics is at the heart of these advances and means that women are starting to be offered bespoke treatments designed to maximise the oncological benefits, whilst minimising the risk of harmful side effects. This is a trend that is set to continue as our understanding of the interplay between genetic and environmental influences on endometrial carcinogenesis increases.
The dramatic rise in endometrial cancer incidence over the last 30 years has emphasised the need for effective strategies for primary disease prevention, which must be targeted at those with the greatest disease risk. Adequately powered clinical trials are urgently needed to identify the most clinically and cost-effective approach to reducing disease incidence and would be supported by many national gynae-oncology groups. Improving the diagnostic pathway for individuals with postmenopausal bleeding to reduce the number requiring invasive testing is a key priority for women and is likely to benefit from ongoing advances in molecular biotechnology. The hunt is still ongoing to identify an ideal biomarker that will enable the accurate diagnosis of those with early-stage endometrial cancer, whilst offering appropriate reassurance to those without the disease. Endometrial cancer treatment in the future is likely to be determined not by traditional histological variables, but rather by the molecular profile of the tumour, with the potential for marked differences in the use of adjuvant treatments across molecular groups. Targeted therapies, including mTOR and PARP inhibitors and antiangiogenic drugs, are currently under investigation for the treatment of advanced and recurrent disease and may well have a role to play in improving survival in the future. Predictive biomarkers used to identify those who will benefit most from these often toxic and expensive therapies will be critical. Attracting funding for endometrial cancer research is essential if we are to continue to make advances in the diagnosis and treatment of the disease at pace.

Author Contributions

Conceptualisation, S.J.K.; investigation, H.B.-R. and S.J.K.; resources, H.B.-R. and S.J.K.; supervision, S.J.K.; writing—original draft, H.B.-R. and S.J.K.; writing—review and editing S.J.K. All authors have read and agreed to the published version of the manuscript.

Funding

H.B.-R is a Cancer Research UK-funded Clinical Research Fellow. Her work is supported by Cancer Grand Challenges (CGCATF-2021/100007).

Conflicts of Interest

The authors have no conflicts of interest to declare.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  2. Cancer Research UK. Uterine Cancer Statistics. Available online: https://www.cancerresearchuk.org/health-professional/cancer-statistics/statistics-by-cancer-type/uterine-cancer#heading-Six (accessed on 17 June 2023).
  3. Gu, B.; Shang, X.; Yan, M.; Li, X.; Wang, W.; Wang, Q.; Zhang, C. Variations in incidence and mortality rates of endometrial cancer at the global, regional, and national levels, 1990–2019. Gynecol. Oncol. 2021, 161, 573–580. [Google Scholar] [CrossRef]
  4. Moss, E.L.; Teece, L.; Darko, N. Uterine cancer mortality and Black women: Time to act. Lancet Oncol. 2023, 24, 586–588. [Google Scholar] [CrossRef]
  5. Clarke, M.A.; Devesa, S.S.; Hammer, A.; Wentzensen, N. Racial and Ethnic Differences in Hysterectomy-Corrected Uterine Corpus Cancer Mortality by Stage and Histologic Subtype. JAMA Oncol. 2022, 8, 895–903. [Google Scholar] [CrossRef] [PubMed]
  6. Ryan, N.A.J.; Glaire, M.A.; Blake, D.; Cabrera-Dandy, M.; Evans, D.G.; Crosbie, E.J. The proportion of endometrial cancers associated with Lynch syndrome: A systematic review of the literature and meta-analysis. Genet. Med. 2019, 21, 2167–2180. [Google Scholar] [CrossRef]
  7. Brown, K.F.; Rumgay, H.; Dunlop, C.; Ryan, M.; Quartly, F.; Cox, A.; Deas, A.; Elliss-Brookes, L.; Gavin, A.; Hounsome, L.; et al. The fraction of cancer attributable to modifiable risk factors in England, Wales, Scotland, Northern Ireland, and the United Kingdom in 2015. Br. J. Cancer 2018, 118, 1130–1141. [Google Scholar] [CrossRef]
  8. Fortner, R.T.; Hüsing, A.; Dossus, L.; Tjønneland, A.; Overvad, K.; Dahm, C.C.; Arveux, P.; Fournier, A.; Kvaskoff, M.; Schulze, M.B.; et al. Theoretical potential for endometrial cancer prevention through primary risk factor modification: Estimates from the EPIC cohort. Int. J. Cancer 2020, 147, 1325–1333. [Google Scholar] [CrossRef] [PubMed]
  9. Kohler, L.N.; Garcia, D.O.; Harris, R.B.; Oren, E.; Roe, D.J.; Jacobs, E.T. Adherence to Diet and Physical Activity Cancer Prevention Guidelines and Cancer Outcomes: A Systematic Review. Cancer Epidemiol. Biomark. Prev. 2016, 25, 1018–1028. [Google Scholar] [CrossRef]
  10. Crosbie, E.J.; Kitson, S.J.; McAlpine, J.N.; Mukhopadhyay, A.; Powell, M.E.; Singh, N. Endometrial cancer. Lancet 2022, 399, 1412–1428. [Google Scholar] [CrossRef]
  11. Kitson, S.J.; Evans, D.G.; Crosbie, E.J. Identifying High-Risk Women for Endometrial Cancer Prevention Strategies: Proposal of an Endometrial Cancer Risk Prediction Model. Cancer Prev. Res. 2017, 10, 1–13. [Google Scholar] [CrossRef] [PubMed]
  12. Luo, J.; Chlebowski, R.T.; Hendryx, M.; Rohan, T.; Wactawski-Wende, J.; Thomson, C.A.; Felix, A.S.; Chen, C.; Barrington, W.; Coday, M.; et al. Intentional Weight Loss and Endometrial Cancer Risk. J. Clin. Oncol. 2017, 35, 1189–1193. [Google Scholar] [CrossRef] [PubMed]
  13. Zhang, X.; Rhoades, J.; Caan, B.J.; Cohn, D.E.; Salani, R.; Noria, S.; Suarez, A.A.; Paskett, E.D.; Felix, A.S. Intentional weight loss, weight cycling, and endometrial cancer risk: A systematic review and meta-analysis. Int. J. Gynecol. Cancer 2019, 29, 1361–1371. [Google Scholar] [CrossRef] [PubMed]
  14. Kitson, S.J.; Aurangzeb, O.; Parvaiz, J.; Lophatananon, A.; Muir, K.R.; Crosbie, E.J. Quantifying the Effect of Physical Activity on Endometrial Cancer Risk. Cancer Prev. Res. 2022, 15, 605–621. [Google Scholar] [CrossRef] [PubMed]
  15. Rubino, D.; Abrahamsson, N.; Davies, M.; Hesse, D.; Greenway, F.L.; Jensen, C.; Lingvay, I.; Mosenzon, O.; Rosenstock, J.; Rubio, M.A.; et al. Effect of Continued Weekly Subcutaneous Semaglutide vs Placebo on Weight Loss Maintenance in Adults With Overweight or Obesity: The STEP 4 Randomized Clinical Trial. JAMA 2021, 325, 1414–1425. [Google Scholar] [CrossRef] [PubMed]
  16. Valladales-Restrepo, L.F.; Sánchez-Ramírez, N.; Usma-Valencia, A.F.; Gaviria-Mendoza, A.; Machado-Duque, M.E.; Machado-Alba, J.E. Effectiveness, persistence of use, and safety of orlistat and liraglutide in a group of patients with obesity. Expert. Opin. Pharmacother. 2023, 24, 535–543. [Google Scholar] [CrossRef] [PubMed]
  17. Melson, E.; Ashraf, U.; Papamargaritis, D.; Davies, M.J. What is the pipeline for future medications for obesity? Int. J. Obes. 2024. ahead of print. [Google Scholar] [CrossRef]
  18. Garvey, W.T.; Batterham, R.L.; Bhatta, M.; Buscemi, S.; Christensen, L.N.; Frias, J.P.; Jódar, E.; Kandler, K.; Rigas, G.; Wadden, T.A.; et al. Two-year effects of semaglutide in adults with overweight or obesity: The STEP 5 trial. Nat. Med. 2022, 28, 2083–2091. [Google Scholar] [CrossRef]
  19. Wadden, T.A.; Tronieri, J.S.; Sugimoto, D.; Lund, M.T.; Auerbach, P.; Jensen, C.; Rubino, D. Liraglutide 3.0 mg and Intensive Behavioral Therapy (IBT) for Obesity in Primary Care: The SCALE IBT Randomized Controlled Trial. Obesity 2020, 28, 529–536. [Google Scholar] [CrossRef]
  20. Pucci, A.; Batterham, R.L. Mechanisms underlying the weight loss effects of RYGB and SG: Similar, yet different. J. Endocrinol. Investig. 2019, 42, 117–128. [Google Scholar] [CrossRef]
  21. Haghighat, N.; Kazemi, A.; Asbaghi, O.; Jafarian, F.; Moeinvaziri, N.; Hosseini, B.; Amini, M. Long-term effect of bariatric surgery on body composition in patients with morbid obesity: A systematic review and meta-analysis. Clin. Nutr. 2021, 40, 1755–1766. [Google Scholar] [CrossRef]
  22. Wilson, R.B.; Lathigara, D.; Kaushal, D. Systematic Review and Meta-Analysis of the Impact of Bariatric Surgery on Future Cancer Risk. Int. J. Mol. Sci. 2023, 24, 6192. [Google Scholar] [CrossRef]
  23. Holmberg, D.; Lagergren, J. What do we know about long-term effects of bariatric surgery? Br. J. Surg. 2022, 110, 1–2. [Google Scholar] [CrossRef]
  24. Allen, N.; Yang, T.O.; Olsson, H. Endometrial cancer and oral contraceptives: An individual participant meta-analysis of 27 276 women with endometrial cancer from 36 epidemiological studies. Lancet Oncol. 2015, 16, 1061–1070. [Google Scholar] [CrossRef]
  25. Faculty of Sexual and Reproductive Healthcare. Overweight, Obesity and Contraception; FSRH: London, UK, 2019. [Google Scholar]
  26. Crosbie, E.J.; Ryan, N.A.J.; Arends, M.J.; Bosse, T.; Burn, J.; Cornes, J.M.; Crawford, R.; Eccles, D.; Frayling, I.M.; Ghaem-Maghami, S.; et al. The Manchester International Consensus Group recommendations for the management of gynecological cancers in Lynch syndrome. Genet. Med. 2019, 21, 2390–2400. [Google Scholar] [CrossRef]
  27. Iversen, L.; Fielding, S.; Lidegaard, Ø.; Hannaford, P.C. Contemporary hormonal contraception and risk of endometrial cancer in women younger than age 50: A retrospective cohort study of Danish women. Contraception 2020, 102, 152–158. [Google Scholar] [CrossRef]
  28. Soini, T.; Hurskainen, R.; Grenman, S.; Maenpaa, J.; Paavonen, J.; Pukkala, E. Cancer risk in women using the levonorgestrel-releasing intrauterine system in Finland. Obstet. Gynecol. 2014, 124, 292–299. [Google Scholar] [CrossRef] [PubMed]
  29. Jareid, M.; Thalabard, J.C.; Aarflot, M.; Bøvelstad, H.M.; Lund, E.; Braaten, T. Levonorgestrel-releasing intrauterine system use is associated with a decreased risk of ovarian and endometrial cancer, without increased risk of breast cancer. Results from the NOWAC Study. Gynecol. Oncol. 2018, 149, 127–132. [Google Scholar] [CrossRef]
  30. Derbyshire, A.E.; Allen, J.L.; Gittins, M.; Lakhiani, B.; Bolton, J.; Shaw, J.; Pemberton, P.W.; Needham, M.; MacKintosh, M.L.; Edmondson, R.J.; et al. PROgesterone Therapy for Endometrial Cancer Prevention in Obese Women (PROTEC) Trial: A Feasibility Study. Cancer Prev. Res. 2021, 14, 263–274. [Google Scholar] [CrossRef]
  31. Bernard, L.; Kwon, J.S.; Simpson, A.N.; Ferguson, S.E.; Sinasac, S.; Pina, A.; Reade, C.J. The levonorgestrel intrauterine system for prevention of endometrial cancer in women with obesity: A cost-effectiveness study. Gynecol. Oncol. 2021, 161, 367–373. [Google Scholar] [CrossRef] [PubMed]
  32. Silva, F.R.; Grande, A.J.; Lacerda Macedo, A.C.; Colonetti, T.; Rocha, M.C.; Rodrigues Uggioni, M.L.; Silva, B.R.; da Rosa, M.I. Meta-Analysis of Breast Cancer Risk in Levonorgestrel-Releasing Intrauterine System Users. Clin. Breast Cancer 2021, 21, 497–508. [Google Scholar] [CrossRef] [PubMed]
  33. Dossus, L.; Rinaldi, S.; Becker, S.; Lukanova, A.; Tjonneland, A.; Olsen, A.; Stegger, J.; Overvad, K.; Chabbert-Buffet, N.; Jimenez-Corona, A.; et al. Obesity, inflammatory markers, and endometrial cancer risk: A prospective case-control study. Endocr. Relat. Cancer 2010, 17, 1007–1019. [Google Scholar] [CrossRef]
  34. Modugno, F.; Ness, R.B.; Chen, C.; Weiss, N.S. Inflammation and endometrial cancer: A hypothesis. Cancer Epidemiol. Biomark. Prev. 2005, 14, 2840–2847. [Google Scholar] [CrossRef]
  35. Verdoodt, F.; Friis, S.; Dehlendorff, C.; Albieri, V.; Kjaer, S.K. Non-steroidal anti-inflammatory drug use and risk of endometrial cancer: A systematic review and meta-analysis of observational studies. Gynecol. Oncol. 2016, 140, 352–358. [Google Scholar] [CrossRef]
  36. Burn, J.; Sheth, H.; Elliott, F.; Reed, L.; Macrae, F.; Mecklin, J.P.; Möslein, G.; McRonald, F.E.; Bertario, L.; Evans, D.G.; et al. Cancer prevention with aspirin in hereditary colorectal cancer (Lynch syndrome), 10-year follow-up and registry-based 20-year data in the CAPP2 study: A double-blind, randomised, placebo-controlled trial. Lancet 2020, 395, 1855–1863. [Google Scholar] [CrossRef] [PubMed]
  37. Kitson, S.; Maskell, Z.; Sivalingam, V.N.; Allen, J.L.; Ali, S.; Burns, S.; Gilmour, K.; Latheef, R.; Slade, R.J.; Pemberton, P.; et al. PRE-surgical Metformin in Uterine Malignancy (PREMIUM): A multi-center, randomized double-blind, placebo-controlled phase 3 trial. Clin. Cancer Res. 2019, 25, 2424–2432. [Google Scholar] [CrossRef] [PubMed]
  38. Clement, N.S.; Oliver, T.R.; Shiwani, H.; Sanner, J.R.; Mulvaney, C.A.; Atiomo, W. Metformin for endometrial hyperplasia. Cochrane Database Syst. Rev. 2017, 10, CD012214. [Google Scholar] [CrossRef] [PubMed]
  39. Chu, D.; Wu, J.; Wang, K.; Zhao, M.; Wang, C.; Li, L.; Guo, R. Effect of metformin use on the risk and prognosis of endometrial cancer: A systematic review and meta-analysis. BMC Cancer 2018, 18, 438. [Google Scholar] [CrossRef]
  40. Janda, M.; Robledo, K.P.; Gebski, V.; Armes, J.E.; Alizart, M.; Cummings, M.; Chen, C.; Leung, Y.; Sykes, P.; McNally, O.; et al. Complete pathological response following levonorgestrel intrauterine device in clinically stage 1 endometrial adenocarcinoma: Results of a randomized clinical trial. Gynecol. Oncol. 2021, 161, 143–151. [Google Scholar] [CrossRef]
  41. Moller, P.; Seppala, T.; Bernstein, I.; Holinski-Feder, E.; Sala, P.; Evans, D.G.; Lindblom, A.; Macrae, F.; Blanco, I.; Sijmons, R.; et al. Cancer incidence and survival in Lynch syndrome patients receiving colonoscopic and gynaecological surveillance: First report from the prospective Lynch syndrome database. Gut 2017, 66, 464–472. [Google Scholar] [CrossRef]
  42. Pfeiffer, R.M.; Park, Y.; Kreimer, A.R.; Lacey, J.V., Jr.; Pee, D.; Greenlee, R.T.; Buys, S.S.; Hollenbeck, A.; Rosner, B.; Gail, M.H.; et al. Risk prediction for breast, endometrial, and ovarian cancer in white women aged 50 y or older: Derivation and validation from population-based cohort studies. PLoS Med. 2013, 10, e1001492. [Google Scholar] [CrossRef]
  43. Husing, A.; Dossus, L.; Ferrari, P.; Tjonneland, A.; Hansen, L.; Fagherazzi, G.; Baglietto, L.; Schock, H.; Chang-Claude, J.; Boeing, H.; et al. An epidemiological model for prediction of endometrial cancer risk in Europe. Eur. J. Epidemiol. 2016, 31, 51–60. [Google Scholar] [CrossRef]
  44. Shi, J.; Kraft, P.; Rosner, B.; Benavente, Y.; Black, A.; Brinton, L.A.; Chen, C.; Clarke, M.A.; Cook, L.S.; Costas, L.; et al. Risk prediction models for endometrial cancer: Development and validation in an international consortium. J. Natl. Cancer Inst. 2023, 115, 552–559. [Google Scholar] [CrossRef]
  45. Kitson, S.J.; Crosbie, E.J.; Evans, D.G.; Lophatananon, A.; Muir, K.R.; Ashcroft, D.; Kontopantelis, E.; Martin, G.P. Predicting risk of endometrial cancer in asymptomatic women (PRECISION): Model development and external validation. BJOG 2023. early view. [Google Scholar] [CrossRef]
  46. Kitson, S.J.; Khan, U.; Crosbie, E.J. Lay and general practitioner attitudes towards endometrial cancer prevention: A cross-sectional study. Fam. Pract. 2023, 2023, cmad076. [Google Scholar] [CrossRef]
  47. van Hanegem, N.; Prins, M.M.; Bongers, M.Y.; Opmeer, B.C.; Sahota, D.S.; Mol, B.W.; Timmermans, A. The accuracy of endometrial sampling in women with postmenopausal bleeding: A systematic review and meta-analysis. Eur. J. Obstet. Gynecol. Reprod. Biol. 2016, 197, 147–155. [Google Scholar] [CrossRef]
  48. Liu, J.; Han, L.; Sun, Q.; Li, Y.; Niyazi, M. Meta-analysis of the diagnostic accuracy of HE4 for endometrial carcinoma. Eur. J. Obstet. Gynecol. Reprod. Biol. 2020, 252, 404–411. [Google Scholar] [CrossRef]
  49. Li, L.M.; Zhu, Y.X.; Zhong, Y.; Su, T.; Fan, X.M.; Xi, Q.; Li, M.Y.; Fu, J.; Tan, H.; Liu, S. Human epididymis protein 4 in endometrial cancer: A meta-analysis. Clin. Chim. Acta 2018, 482, 215–223. [Google Scholar] [CrossRef]
  50. Knific, T.; Osredkar, J.; Smrkolj, Š.; Tonin, I.; Vouk, K.; Blejec, A.; Frković Grazio, S.; Rižner, T.L. Novel algorithm including CA-125, HE4 and body mass index in the diagnosis of endometrial cancer. Gynecol. Oncol. 2017, 147, 126–132. [Google Scholar] [CrossRef] [PubMed]
  51. Bolivar, A.M.; Luthra, R.; Mehrotra, M.; Chen, W.; Barkoh, B.A.; Hu, P.; Zhang, W.; Broaddus, R.R. Targeted next-generation sequencing of endometrial cancer and matched circulating tumor DNA: Identification of plasma-based, tumor-associated mutations in early stage patients. Mod. Pathol. 2019, 32, 405–414. [Google Scholar] [CrossRef] [PubMed]
  52. Beinse, G.; Borghese, B.; Métairie, M.; Just, P.A.; Poulet, G.; Garinet, S.; Parfait, B.; Didelot, A.; Bourreau, C.; Agueeff, N.; et al. Highly Specific Droplet-Digital PCR Detection of Universally Methylated Circulating Tumor DNA in Endometrial Carcinoma. Clin. Chem. 2022, 68, 782–793. [Google Scholar] [CrossRef] [PubMed]
  53. Fan, X.; Zou, X.; Liu, C.; Cheng, W.; Zhang, S.; Geng, X.; Zhu, W. MicroRNA expression profile in serum reveals novel diagnostic biomarkers for endometrial cancer. Biosci. Rep. 2021, 41, BSR20210111. [Google Scholar] [CrossRef]
  54. Nair, N.; Camacho-Vanegas, O.; Rykunov, D.; Dashkoff, M.; Camacho, S.C.; Schumacher, C.A.; Irish, J.C.; Harkins, T.T.; Freeman, E.; Garcia, I.; et al. Genomic Analysis of Uterine Lavage Fluid Detects Early Endometrial Cancers and Reveals a Prevalent Landscape of Driver Mutations in Women without Histopathologic Evidence of Cancer: A Prospective Cross-Sectional Study. PLoS Med. 2016, 13, e1002206. [Google Scholar] [CrossRef]
  55. Wang, Y.; Li, L.; Douville, C.; Cohen, J.D.; Yen, T.T.; Kinde, I.; Sundfelt, K.; Kjær, S.K.; Hruban, R.H.; Shih, I.M.; et al. Evaluation of liquid from the Papanicolaou test and other liquid biopsies for the detection of endometrial and ovarian cancers. Sci. Transl. Med. 2018, 10, eaap8793. [Google Scholar] [CrossRef]
  56. Pelegrina, B.; Paytubi, S.; Marin, F.; Martínez, J.M.; Carmona, Á.; Frias-Gomez, J.; Peremiquel-Trillas, P.; Dorca, E.; Zanca, A.; López-Querol, M.; et al. Evaluation of somatic mutations in cervicovaginal samples as a non-invasive method for the detection and molecular classification of endometrial cancer. EBioMedicine 2023, 94, 104716. [Google Scholar] [CrossRef]
  57. Evans, I.; Reisel, D.; Jones, A.; Bajrami, A.; Nijjar, S.; Solangon, S.A.; Arora, R.; Redl, E.; Schreiberhuber, L.; Ishaq-Parveen, I.; et al. Performance of the WID-qEC test versus sonography to detect uterine cancers in women with abnormal uterine bleeding (EPI-SURE): A prospective, consecutive observational cohort study in the UK. Lancet Oncol. 2023, 24, 1375–1386. [Google Scholar] [CrossRef] [PubMed]
  58. O’Flynn, H.; Ryan, N.A.J.; Narine, N.; Shelton, D.; Rana, D.; Crosbie, E.J. Diagnostic accuracy of cytology for the detection of endometrial cancer in urine and vaginal samples. Nat. Commun. 2021, 12, 952. [Google Scholar] [CrossRef]
  59. Burke, T.W.; Levenback, C.; Tornos, C.; Morris, M.; Wharton, J.T.; Gershenson, D.M. Intraabdominal lymphatic mapping to direct selective pelvic and paraaortic lymphadenectomy in women with high-risk endometrial cancer: Results of a pilot study. Gynecol. Oncol. 1996, 62, 169–173. [Google Scholar] [CrossRef] [PubMed]
  60. Nagar, H.; Wietek, N.; Goodall, R.J.; Hughes, W.; Schmidt-Hansen, M.; Morrison, J. Sentinel node biopsy for diagnosis of lymph node involvement in endometrial cancer. Cochrane Database Syst. Rev. 2021, 6, CD013021. [Google Scholar] [CrossRef] [PubMed]
  61. Frumovitz, M.; Plante, M.; Lee, P.S.; Sandadi, S.; Lilja, J.F.; Escobar, P.F.; Gien, L.T.; Urbauer, D.L.; Abu-Rustum, N.R. Near-infrared fluorescence for detection of sentinel lymph nodes in women with cervical and uterine cancers (FILM): A randomised, phase 3, multicentre, non-inferiority trial. Lancet Oncol. 2018, 19, 1394–1403. [Google Scholar] [CrossRef] [PubMed]
  62. 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. Radiother. Oncol. 2021, 154, 327–353. [Google Scholar] [CrossRef]
  63. Abu-Rustum, N.R.; Yashar, C.M.; Bradley, K.; Campos, S.M.; Chino, J.; Chon, H.S.; Chu, C.; Cohn, D.; Crispens, M.A.; Damast, S.; et al. NCCN Guidelines® Insights: Uterine Neoplasms, Version 3.2021. J. Natl. Compr. Cancer Netw. 2021, 19, 888–895. [Google Scholar] [CrossRef]
  64. Frost, J.A.; Webster, K.E.; Bryant, A.; Morrison, J. Lymphadenectomy for the management of endometrial cancer. Cochrane Database Syst. Rev. 2017, 10, CD007585. [Google Scholar] [CrossRef]
  65. Cusimano, M.C.; Simpson, A.N.; Dossa, F.; Liani, V.; Kaur, Y.; Acuna, S.A.; Robertson, D.; Satkunaratnam, A.; Bernardini, M.Q.; Ferguson, S.E.; et al. Laparoscopic and robotic hysterectomy in endometrial cancer patients with obesity: A systematic review and meta-analysis of conversions and complications. Am. J. Obstet. Gynecol. 2019, 221, 410–428.e419. [Google Scholar] [CrossRef]
  66. Lim, Y.H.; Dagher, C.; Abu-Rustum, N.R.; Mueller, J.J.; Sonoda, Y.; Zivanovic, O.; Broach, V.; Leitao, M.M. Oncologic outcomes of robot-assisted laparoscopy versus conventional laparoscopy for the treatment of apparent early-stage endometrioid adenocarcinoma of the uterus. Gynecol. Oncol. 2023, 179, 152–157. [Google Scholar] [CrossRef] [PubMed]
  67. Fu, H.; Zhang, J.; Zhao, S.; He, N. Survival outcomes of robotic-assisted laparoscopy versus conventional laparoscopy and laparotomy for endometrial cancer: A systematic review and meta-analysis. Gynecol. Oncol. 2023, 174, 55–67. [Google Scholar] [CrossRef] [PubMed]
  68. Moss, E.L.; Sarhanis, P.; Ind, T.; Smith, M.; Davies, Q.; Zecca, M. Impact of Obesity on Surgeon Ergonomics in Robotic and Straight-Stick Laparoscopic Surgery. J. Minim. Invasive Gynecol. 2020, 27, 1063–1069. [Google Scholar] [CrossRef] [PubMed]
  69. 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. [Google Scholar] [CrossRef] [PubMed]
  70. Albright, B.B.; Monuszko, K.A.; Kaplan, S.J.; Davidson, B.A.; Moss, H.A.; Huang, A.B.; Melamed, A.; Wright, J.D.; Havrilesky, L.J.; Previs, R.A. Primary cytoreductive surgery for advanced stage endometrial cancer: A systematic review and meta-analysis. Am. J. Obstet. Gynecol. 2021, 225, 237.e1–237.e4. [Google Scholar] [CrossRef] [PubMed]
  71. Huang, A.B.; Wu, J.; Chen, L.; Albright, B.B.; Previs, R.A.; Moss, H.A.; Davidson, B.A.; Havrilesky, L.J.; Melamed, A.; Wright, J.D. Neoadjuvant chemotherapy for advanced stage endometrial cancer: A systematic review. Gynecol. Oncol. Rep. 2021, 38, 100887. [Google Scholar] [CrossRef] [PubMed]
  72. Berek, J.S.; Matias-Guiu, X.; Creutzberg, C.; Fotopoulou, C.; Gaffney, D.; Kehoe, S.; Lindemann, K.; Mutch, D.; Concin, N.; Endometrial Cancer Staging Subcommittee, FIGO Women’s Cancer Committee. FIGO staging of endometrial cancer: 2023. J. Gynecol. Oncol. 2023, 34, e85. [Google Scholar] [CrossRef]
  73. The Cancer Genome Atlas Research Network; Kandoth, C.; Schultz, N.; Cherniack, A.D.; Akbani, R.; Liu, Y.; Shen, H.; Robertson, A.G.; Pashtan, I.; Shen, R.; et al. Integrated genomic characterization of endometrial carcinoma. Nature 2013, 497, 67–73. [Google Scholar] [CrossRef]
  74. Clinicaltrials.gov. Refining Adjuvant Treatment in Endometrial Cancer Based on Molecular Features (RAINBO). 2024. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT05255653 (accessed on 29 February 2024).
  75. Stelloo, E.; Bosse, T.; Nout, R.A.; MacKay, H.J.; Church, D.N.; Nijman, H.W.; Leary, A.; Edmondson, R.J.; Powell, M.E.; Crosbie, E.J.; et al. Refining prognosis and identifying targetable pathways for high-risk endometrial cancer; a TransPORTEC initiative. Mod. Pathol. 2015, 28, 836–844. [Google Scholar] [CrossRef]
  76. 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. [Google Scholar] [CrossRef]
  77. León-Castillo, A.; de Boer, S.M.; Powell, M.E.; Mileshkin, L.R.; Mackay, H.J.; Leary, A.; Nijman, H.W.; Singh, N.; Pollock, P.M.; Bessette, P.; et al. Molecular Classification of the PORTEC-3 Trial for High-Risk Endometrial Cancer: Impact on Prognosis and Benefit From Adjuvant Therapy. J. Clin. Oncol. 2020, 38, 3388–3397. [Google Scholar] [CrossRef]
  78. Llosa, N.J.; Cruise, M.; Tam, A.; Wicks, E.C.; Hechenbleikner, E.M.; Taube, J.M.; Blosser, R.L.; Fan, H.; Wang, H.; Luber, B.S.; et al. The vigorous immune microenvironment of microsatellite instable colon cancer is balanced by multiple counter-inhibitory checkpoints. Cancer Discov. 2015, 5, 43–51. [Google Scholar] [CrossRef] [PubMed]
  79. O’Malley, D.M.; Bariani, G.M.; Cassier, P.A.; Marabelle, A.; Hansen, A.R.; De Jesus Acosta, A.; Miller, W.H.; Safra, T.; Italiano, A.; Mileshkin, L.; et al. Pembrolizumab in Patients with Microsatellite Instability-High Advanced Endometrial Cancer: Results From the KEYNOTE-158 Study. J. Clin. Oncol. 2022, 40, 752–761. [Google Scholar] [CrossRef] [PubMed]
  80. 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. [Google Scholar] [CrossRef]
  81. Dioun, S.; Chen, L.; Melamed, A.; Gockley, A.; St Clair, C.M.; Hou, J.Y.; Khoury-Collado, F.; Hur, C.; Elkin, E.; Accordino, M.; et al. Dostarlimab for recurrent mismatch repair-deficient endometrial cancer: A cost-effectiveness study. BJOG 2023, 130, 214–221. [Google Scholar] [CrossRef] [PubMed]
  82. Makker, V.; Colombo, N.; Casado Herráez, A.; Santin, A.D.; Colomba, E.; Miller, D.S.; Fujiwara, K.; Pignata, S.; Baron-Hay, S.; Ray-Coquard, I.; et al. Lenvatinib plus Pembrolizumab for Advanced Endometrial Cancer. N. Engl. J. Med. 2022, 386, 437–448. [Google Scholar] [CrossRef] [PubMed]
  83. Mirza, M.R.; Chase, D.M.; Slomovitz, B.M.; dePont Christensen, R.; Novák, Z.; Black, D.; Gilbert, L.; Sharma, S.; Valabrega, G.; Landrum, L.M.; et al. Dostarlimab for Primary Advanced or Recurrent Endometrial Cancer. N. Engl. J. Med. 2023, 388, 2145–2158. [Google Scholar] [CrossRef] [PubMed]
  84. Makker, V.; Taylor, M.H.; Aghajanian, C.; Cohn, A.L.; Brose, M.S.; Simone, C.D.; Cao, Z.A.; Suttner, L.; Loboda, A.; Cristescu, R.; et al. Evaluation of potential biomarkers for lenvatinib plus pembrolizumab among patients with advanced endometrial cancer: Results from Study 111/KEYNOTE-146. J. Immunother. Cancer 2024, 12, e007929. [Google Scholar] [CrossRef]
  85. Behnamfar, F.; Ghahiri, A.; Tavakoli, M. Levonorgestrel-releasing intrauterine system (Mirena) in compare to medroxyprogesterone acetate as a therapy for endometrial hyperplasia. J. Res. Med. Sci. 2014, 19, 686–690. [Google Scholar] [PubMed]
  86. Dolapcioglu, K.; Boz, A.; Baloglu, A. The efficacy of intrauterine versus oral progestin for the treatment of endometrial hyperplasia. A prospective randomized comparative study. Clin. Exp. Obstet. Gynecol. 2013, 40, 122–126. [Google Scholar] [PubMed]
  87. Britton, H.; Huang, L.; Lum, A.; Leung, S.; Shum, K.; Kale, M.; Burleigh, A.; Senz, J.; Yang, W.; McConechy, M.; et al. Molecular classification defines outcomes and opportunities in young women with endometrial carcinoma. Gynecol. Oncol. 2019, 153, 487–495. [Google Scholar] [CrossRef]
  88. Gunderson, C.C.; Fader, A.N.; Carson, K.A.; Bristow, R.E. Oncologic and reproductive outcomes with progestin therapy in women with endometrial hyperplasia and grade 1 adenocarcinoma: A systematic review. Gynecol. Oncol. 2012, 125, 477–482. [Google Scholar] [CrossRef] [PubMed]
  89. Barr, C.E.; Ryan, N.A.J.; Derbyshire, A.E.; Wan, Y.L.; MacKintosh, M.L.; McVey, R.J.; Bolton, J.; Fitzgerald, C.; Awad, D.; Slade, R.J.; et al. Weight Loss During Intrauterine Progestin Treatment for Obesity-associated Atypical Hyperplasia and Early-Stage Cancer of The Endometrium. Cancer Prev. Res. 2021, 14, 1041–1050. [Google Scholar] [CrossRef]
Table 1. Prognostic and therapeutic implications of the molecular classification of endometrial cancers.
Table 1. Prognostic and therapeutic implications of the molecular classification of endometrial cancers.
Molecular GroupIdentifying FeaturesSurrogate MarkerPredominant Endometrial Cancer HistotypePrognosis (Progression Free Survival at 5 Years)Therapeutic Implication
POLEmutVery high mutational load POLE exonuclease domain mutationAll histotypes with the exception of serous carcinomasExcellent (92–100%)Adjuvant treatment may not be required given excellent prognosis
MMRdHigh mutational loadLoss of MLH1, MSH2, MSH6 and/or PMS2 expressionPredominately endometrioid carcinomasIntermediate (80–90%)Limited benefit from chemotherapy. Improved response to immunotherapy.
p53abnLow mutational load, high copy number variationsAbnormal p53 expressionSerous, high grade endometrioidPoor (50%)Benefit from concurrent chemo-radiotherapy. PARP inhibitor therapy may also be effective
NSMPLow mutational load, low copy number variationsAbsence of the other markersLow grade endometrioidHeterogeneous but overall considered intermediate (75–80%)May benefit from hormonal treatment given frequent ER+ and PR+
Table 2. Indications for immunotherapy and predictive biomarkers of response. The underlined terms signal the different licensed indications for single agent or combination therapy.
Table 2. Indications for immunotherapy and predictive biomarkers of response. The underlined terms signal the different licensed indications for single agent or combination therapy.
Immunotherapy
Regime
Licensed IndicationCurrent Predictive
Biomarker
Predictive Biomarkers under Investigation
PembrolizumabAdvanced or recurrent MSI-H or MMRd endometrial cancer with disease progression following previous systemic treatment and not suitable for curative surgery or radiotherapyMSI-H and MMRdTumoural immune profile (CD4, CD8 cell populations)
DostarlimabSingle agent-advanced or recurrent MSI-H or MMRd endometrial cancer with disease progression following previous systemic treatment and not suitable for curative surgery or radiotherapy
In combination with carboplatin and paclitaxel for primary advanced or recurrent MSI-H or MMRd endometrial cancer
MSI-H and MMRdTumour mutational burden, PD-L1 expression, ctDNA
Pembrolizumab plus LenvatinibAdvanced or recurrent endometrial cancer, regardless of MMR and MSI status, with disease progression on or following systemic treatment and not suitable for curative surgery or radiotherapyNoneTumour mutational burden, T cell-inflammation gene signature
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Baker-Rand, H.; Kitson, S.J. Recent Advances in Endometrial Cancer Prevention, Early Diagnosis and Treatment. Cancers 2024, 16, 1028. https://doi.org/10.3390/cancers16051028

AMA Style

Baker-Rand H, Kitson SJ. Recent Advances in Endometrial Cancer Prevention, Early Diagnosis and Treatment. Cancers. 2024; 16(5):1028. https://doi.org/10.3390/cancers16051028

Chicago/Turabian Style

Baker-Rand, Holly, and Sarah J. Kitson. 2024. "Recent Advances in Endometrial Cancer Prevention, Early Diagnosis and Treatment" Cancers 16, no. 5: 1028. https://doi.org/10.3390/cancers16051028

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop