Abstract
Background
Fifteen percent of patients with endometrial cancer (EC) have advanced stage disease or develop a recurrence. Progestins have been applied as systemic treatment for decades, but there is limited evidence on response prediction with biomarkers and toxicity.
Objectives
To review the response and toxicity of progestin therapy and stratify response to progesterone receptor (PR) expression and tumour grade.
Search strategy
We used the search terms ‘Endometrial cancer’, ‘Progestins’, ‘Disease progression’, ‘Recurrence’ and related terms in Pubmed, Embase and Cochrane databases.
Selection criteria
Studies on patients with advanced stage or recurrent EC treated with progestin monotherapy were included. Studies on adjuvant therapy, with fewer than ten cases and with sarcoma histology were excluded.
Data collection and analysis
Evaluation for bias was performed with the Revised Cochrane RoB2 tool for randomised studies and the ROBINS‐I tool for non‐randomised studies. A random effects meta‐analysis was performed with the overall response rate (ORR), clinical benefit rate and toxicity as primary outcome measures.
Main results
Twenty‐six studies (1639 patients) were included. The ORR of progestin therapy was 30% (95% CI 25–36), the clinical benefit rate was 52% (95% CI 42–61). In PR‐positive EC, the ORR was 55%, compared with 12% in PR‐negative disease (risk difference 43%, 95% CI 15–71). Severe toxicity occurred in 6.5%.
Conclusions
Progestin therapy is a viable treatment option in patients with advanced stage and recurrent EC with low toxicity and high ORR in PR‐positive disease. The role of PR expression in relation to progression‐free survival and overall survival is unclear.
Keywords: advanced stage, endometrial cancer, medroxyprogesterone acetate, megestrol acetate, meta‐analysis, progestin therapy, recurrence, systematic review
1. INTRODUCTION
Progestins have a multitude of applications in gynaecological practice. 1 , 2 , 3 , 4 , 5 Aside from their use for menstrual disorders, endometriosis and prevention of preterm birth, progestin therapy can be applied in women with endometrial cancer (EC) for fertility preservation and as palliative treatment in advanced stage or recurrent disease. 6 Approximately 15% of EC patients present either with advanced stage disease or recurrence; for these patients prognosis is poor and curative treatment options are limited. 7 , 8 , 9 , 10 , 11 Hormonal therapy and chemotherapy are the most frequently used palliative systemic treatment options. 12 First‐line chemotherapy consists of carboplatin and paclitaxel and is beneficial in 50–60% of patients. 13 , 14 , 15 , 16 However, this therapy is associated with substantial toxicity including sensory neuropathy in 20% of patients. 13 , 16 Hormonal therapy on the other hand yields response rates of 20–40% in unselected populations, but with limited toxicity. 17 , 18
Hormonal therapy was introduced in the 1960s when progestins were shown to have antiproliferative properties that counteract the effects of estrogens in EC. 19 Today, hormonal therapy is increasingly used, mostly as progestins, with other drugs like selective estrogen receptor modulators and degraders (i.e. tamoxifen) and aromatase inhibitors being reserved as second‐line treatment options. 8 , 20 The response to hormonal therapy can be predicted by the presence of estrogen receptors (ER) and progesterone receptors (PR) in tumour cells. The ER and PR serve as mediators of intracellular estrogen and progestin action, respectively, and are expressed by endometrial cells. 21 In advanced or recurrent EC, PR expression is frequently lost and, as a result, sensitivity to progestins might be lower. 22 , 23 However, progestin therapy efficacy has mainly been studied in unselected populations. A limited number of studies have evaluated response to progestin therapy stratified by PR status, but response rates vary substantially and implications for clinical practice are therefore unclear. 17 , 24 In addition, previously performed reviews have summarised disease outcome with hormonal therapy, but not focused specifically on progestin therapy. 7 , 25 Inclusion of less active hormonal therapies in these reviews might underestimate the effect of progestin therapy. 26 , 27 Finally, toxicity has not yet been systematically reviewed. 28 Because patients with palliative treatment of EC are often elderly with significant comorbidities, it is essential to have insight into toxicity to ascertain optimal application of progestins in this group of patients. Performance of a systematic review and meta‐analysis might provide more robust evidence that facilitates improved treatment selection for systemic palliative therapy in EC.
2. OBJECTIVES
The aim of this study was to systematically review the efficacy and toxicity of progestin therapy in advanced stage and recurrent EC and investigate the value of predictive biomarkers.
3. METHODS
3.1. Eligibility criteria, information sources, search strategy
This review was performed in accordance with the Preferred Reporting Items for Systematic Reviews and Meta‐Analyses (PRISMA) and Meta‐analysis Of Observational Studies in Epidemiology (MOOSE) guidelines and registered with PROSPERO (CRD42018089801) in March 2018. 29 , 30 , 31 The research question was drafted according to the PICO framework: patients with advanced stage (FIGO Stage III–IV) or recurrent EC were eligible, the intervention was progestin therapy, the comparison was any other treatment or no treatment and the primary outcomes were overall response rate, clinical benefit rate and toxicity. Exclusion criteria included studies consisting of case series with fewer than ten participants, studies on endometrial stroma sarcoma and studies on adjuvant or combined treatment of different types of hormones or hormonal treatment with other therapy. Further exclusion criteria were languages other than English, Dutch or German, systematic reviews and non‐full text availability. Studies that did not report all outcome measures of interest were included in this review for the reported outcomes only.
3.2. Search strategy
An electronic literature search for eligible studies was performed in the databases PubMed, Embase and the Cochrane database of Systematic Reviews from inception to 31 December 2020. The citation lists of included studies were checked to identify further reports of trials. The search terms were ‘Endometrial cancer’, ‘Progestins’ and outcome measures ‘Disease progression’, ‘Survival’ and ‘Recurrence’ and related terms. The complete search string is shown in Table S1.
3.3. Primary and secondary outcome measures
The primary outcomes were overall response rate (ORR), clinical benefit rate (CBR) and toxicity. The ORR was defined as the proportion of patients with a complete or partial response. The CBR was defined as the proportion of patients with a response and with stable disease. Toxicity was defined as any adverse reaction that could be related to the applied hormonal drug in accordance with the protocol of the included studies. The secondary outcomes were overall survival (OS) and progression‐free survival (PFS).
3.4. Study selection
For the selection of eligible studies, the systematic review tool rayyan was used. 32 After exclusion of duplicates, studies were screened based on title and abstract by two of three reviewers (WW, PB and JP). Potential eligible studies were screened based on the complete text of the study. Discrepancies were resolved in consensus meetings of the three reviewers.
3.5. Data extraction
The following patient characteristics were extracted from the studies using a standardised protocol: age, tumour stage and grade, PR status (PR‐positive or PR‐negative), histology, (previous) treatment, duration of study intervention, ORR, CBR, OS, PFS and toxicity. Authors of included articles were contacted if additional data were required.
3.6. Assessment of risk of bias
The quality of a study was appraised using the Revised Cochrane risk‐of‐bias tool for randomised trials (RoB2) for randomised studies and the ROBINS‐I risk‐of‐bias tool for non‐randomised trials. 33 , 34 Two authors (WW and PB) applied the risk‐of‐bias tools independently, followed by a consensus meeting to discuss and solve differences. In case of discrepancies, consensus was reached after discussion with the third reviewer (JP).
3.7. Data synthesis
The meta‐analyses for ORR and CBR were conducted with a random effects model. Between‐study heterogeneity was assessed with the restricted maximum likelihood estimator for τ2, and expressed as I 2. 29 Publication bias was explored with funnel plots and Egger's test. 35 Sensitivity analyses were performed to evaluate any possible biases in the results caused by studies with high risk of bias, studies published before the year 2000, studies that did not apply standardised evaluation criteria and studies that applied other progestin therapies than the currently used megestrol acetate (MA) and medroxyprogesterone acetate (MPA). Subgroup analysis was performed based on the presence of PR and tumour grade (grade 1–2 versus grade 3 and grade 1–2 versus grade 3–4 for studies that applied the tumour grading system as described by Broder). 36 Response was not stratified by ER status because progestin interacts mainly with PR to exert its biological function and a recent report showed that PR is superior to ER in predicting response to hormonal therapy. 37 The PR‐status definition was adopted from the cutoff values defined by the individual studies. The association between response and biomarker presence or absence was expressed as risk difference and analysed with Mantel–Haenszel method with Hartung–Knapp adjustment for a random effects model. Average scores were used to describe the prevalence of overall and severe toxicity. OS and PFS were expressed in ranges stratified by response and PR status if available. The analyses were conducted with the statistical software R, version 3.6.3 (2020), and packages meta (version 4.11‐0), metafor (version 2.1‐0), and mixmeta (version 1.0.8). 38 , 39 , 40
Patients were not involved in the execution of this review. No core outcome set was used in this research.
4. RESULTS
4.1. Study selection
The literature search yielded 7865 records based on application of the search string and screening of citations of eligible studies (Figure 1). After removal of duplicates and screening for title and abstract, 181 records were assessed for eligibility. A total of 155 records were excluded because of: case series with fewer than ten cases (n = 20), review/comment or guideline (n = 33) and wrong population or treatment (n = 102) including studies on fertility preservation and adjuvant progestin therapy mainly. Twenty‐six studies were included in this review.
FIGURE 1.

PRISMA flow diagram of selection process of included studies
4.2. Study characteristics
The 26 included studies comprised a total of 1639 patients. Four studies (15.3% of included studies) were randomised controlled trials. 17 , 41 , 42 , 43 One trial compared low‐dose and high‐dose MPA, the second trial compared MA and irosustat (a steroid sulphatase inhibitor), the third compared MA and combined treatment with MA and tamoxifen and the fourth trial compared MPA and tamoxifen. Out of the other studies, ten were prospective (38.5%) and 12 were retrospective (46.2%). The studies were published between the years 1961 and 2017 and 21 (81%) of them were published before the year 2000. The characteristics of the included studies are described in Table S2. Patients in the included studies had a mean age that ranged from 60 to 79 years. Different types and dosages of progestins were applied: treatment with either MA or MPA was applied exclusively in 13 studies (50.0%).
4.3. Risk of bias of included studies
The quality assessment showed a moderate risk of bias for 17 studies (65.3%) and a high risk of bias for nine studies (34.7%, Table S3). None of the studies had a low risk of bias.
4.4. Overall response rates and clinical benefit rate
All studies with the exception of one, reported ORR to progestin therapy, resulting in a total number of 1611 included patients. 44 The ORR was 30% (95% CI 25–36%) with substantial heterogeneity (I 2 79%, Figure 2A). The funnel plot can be found in Figure S1(A). The reported durations of progestin therapy ranged from 18 days to 108 months and the reported duration of response was between 3 and 95 months. CBR was available in 14 studies and 1017 patients and was 52% (95% CI 42–61, I 2 81%, Figure 2B), For the funnel plot see Figure S1(B). Sensitivity analyses for ORR and CBR are shown in Figures S2 and S3, respectively. Among moderate‐risk studies, the ORR was 29% (95% CI 22–36%) and the CBR was 50% (95% CI 40–59%). ORR and CBR were similar for studies published before and after the year 2000. In 19 of 25 studies that reported ORR or CBR, a standardised response scale was applied: in two studies RECIST criteria were used, in three studies GOG response criteria were applied and in one study ECOG response criteria were used. In the other studies, the response criteria ranged from any decrease in tumour size to a complete response, or were not specified (n = 3). The minimally required duration of effect for response was given by six other studies ranging from 4 weeks to 3 months. ORR was lower in studies with standardised response criteria (27% 95% CI 21–33%) compared with studies without defined response criteria (45%) (95% CI 34–56%). There was no difference between studies that applied exclusively MA or MPA compared with other progestins.
FIGURE 2.

Meta‐analysis of overall response rate (A) and clinical benefit rate (B) to progestin therapy.
The PR expression status was reported by seven studies. In two studies, ORR was not reported stratified by PR status leaving five studies and 201 patients. 17 , 24 , 42 , 45 , 46 , 47 , 48 PR status was based on primary tumour tissue in two studies and metastatic tumour lesions in three studies (Figure 3). The method for PR analysis was the ligand‐binding assay with different cutoff values in all included studies. As shown in Figure 3(A), the pooled difference in ORR between PR‐positive and PR‐negative tumours was 43% (95% CI 15–71%, I 2 53%). The ORR for PR‐positive tumours was 55.4%, while PR‐negative tumours had an ORR of 12.2%. CBR stratified by PR status was not reported in any of the included studies. The ORR to progestin therapy stratified by tumour grade was reported by 12 studies including 915 patients (Figure 3B). The pooled difference in ORR between low‐grade and high‐grade tumours was 19% (95% CI 15–24%, I 2 = 0%). Low tumour grade was associated with an ORR of 29.1% whereas high tumour grade yielded an ORR of 9.7%.
FIGURE 3.

Meta‐analysis of overall response rate in subgroup stratified by progesterone receptor status (A) and tumour grade (B). *All available studies used ligand‐binding assays instead of presently used immunohistochemical analysis methods. In the study of Yunokawa and Pautier, response was stratified by PR status, but raw data were not published nor available on request. Abbreviations: fmol, femtomole; high grade, tumour grade 3 (and grade 4 of Broder's grading system); low grade, tumour grade 1–2; PR, progesterone receptor.
4.4.1. Toxicity
Treatment‐related toxicity was reported by 11 studies (Table 1). Adverse effects to progestin treatment occurred in 28.8% of patients and these effects were mainly mild. Severe toxicity was reported by 6.5% of all patients (range between individual studies: 0–31.0%) with the highest severe toxicities reported in the study by Lentz et al., in which megestrol acetate was used at the high dosage of 800 mg/day. 18 Thromboembolic processes were the most common severe adverse effect, occurring in 2.5% of patients.
TABLE 1.
Toxicity for progestin therapy
| Study | N | Progestin type and dose | Total toxicity (%) | Severe toxicity | |||
|---|---|---|---|---|---|---|---|
| Thrombo‐embolism (%) | Weight gain (%) | Other (%) | Total severe toxicity (%) | ||||
| Anderson 59 | 20 | MPA 300 mg/day | No toxicity observed | – | – | – | 0 |
| Karagol 49 | 16 |
MA 160 mg/day MPA 500 mg/day |
Only severe toxicity reported | – | – | – | 0 |
| Kelley 19 | 21 | HPC, Progesterone in oil 150–1000 mg/week | 5 (23.8) | 1 (4.8) | 1 (4.8) | 2 (9.6) | 4 (19.0) |
| Lentz 18 | 58 | MA 800 mg/day |
40 (69.0) |
4 (6.9) | 3 (5.2) | 11 (17.6) | 18 (31.0) |
| Malkasian 60 | 75 | MA, HPC Medrogestone: different dosages | Only severe toxicity reported | – | – | – | 0 |
| Morrison 61 | 16 | Dimethisterone 300 mg/day | No toxicity observed | – | – | – | 0 |
| Pandya 41 | 20 | MA 160 mg/day | 8 (40.0) | – | – | – | 0 |
| Pautier 42 | 35 | MA 160 mg/day | 14 (40.0) | 2 (5.7) | – | 1 (2.9) | 3 (8.6) |
| Rendina 43 | 48 | MPA 1 g/week | 48 (100.0) | – | – | – | 0 |
| Rozier 62 | 44 |
MPA 250 mg/week MA 40 mg/day Medrogestone 600 mg/day |
Only severe toxicity reported | – | 2 (4.5) | 4 (9) | 6 (13.6) |
| Thigpen 17 | 296 | MPA 200 mg/day and 1000 mg/day | 33 (11.1) | 9 (3.0) | – | 2 (0.7) | 11 (3.7) |
| Total (%) | 649 | 148 (28.8) a | 16 (2.5) | 6 (0.9) | 20(3.1) | 42 (6.5) | |
Abbreviations: HPC, hydroxyprogesterone‐caproate; MA, megestrol‐acetate; MPA, medroxyprogesterone‐acetate.
Studies that did not report total toxicity were excluded from this analysis.
4.4.2. Overall survival
Follow up was reported in 4 of the 26 included studies. 44 , 48 , 49 , 50 Data on OS was available in 14 studies. The OS ranged between 2.8 and 57.0 months, depending on initial response to progestins. In responding patients, the range in OS was between 7.0 and 59.8 months, in patients with stable disease, the range in OS was 6.3–42.0 months and for patients with progressive disease, OS ranged from 2.8 to 9.1 months. OS stratified by PR status was investigated in two studies. 17 , 45 Patients with PR‐positive tumours had OS that ranged from 9.0 to 12.1 months, whereas patients with PR‐negative tumours had OS ranging from 6.8 to 7.3 months.
Data on PFS was available from seven studies and was stratified by neither response status nor PR status. The PFS ranged from 2.5 to 10 months.
5. DISCUSSION
5.1. Main findings
In this systematic review and meta‐analysis we have found an overall response rate to progestin treatment of 30%, and the clinical benefit rate was 52%. Progesterone receptor expression was associated with an ORR of 55.4% in PR‐positive EC and 12.2% in PR‐negative EC. The impact of PR expression on PFS and OS is unknown. Severe toxicity was reported in 6.5% of patients, among which thromboembolic processes occurred in 2.5% of patients. The overall quality of the included studies was moderate to low and the type and duration of progestin treatment as well as response evaluation varied substantially between studies.
5.2. Comparison with existing literature
Progestin therapy has been the subject of three earlier systematic reviews. In the Cochrane review performed by Kokka et al., 7 progestin therapy for advanced and recurrent EC was found to have no statistically significant impact on OS. However, the Cochrane review included studies on adjuvant progestin therapy without reported ORR, hence results cannot be compared to our review. The review by Ethier et al. 28 reported an ORR of 23.3% and CBR of 45.8%, which is slightly lower when compared with our findings. This is probably explained by the inclusion of more recent studies with higher ORR in our review. In the review by Decruze et al., 25 the ORR was 23% in line with results of Ethier et al. 28 Compared with the reviews of Decruze and Ethier and their colleagues, our review provides updated information on the efficacy of progestin therapy and the predictive value of PR, tumour grade and toxicity data.
The presence both of ER and of PR can be used as a predictive biomarker. As PR expression is more relevant, we focused on PR expression in this review. 37 , 51 Currently, immunohistochemical (IHC) staining analysis is the reference standard to determine expression of ER and PR. Only two studies in this review used IHC analysis for PR and in these studies, response was not stratified by PR status. 42 , 48 Upon request, these data could unfortunately not be made available for this review. As a result, only studies with an older ligand‐binding assay for PR expression were included. 17 , 24 , 45 , 46 , 47 Ligand‐binding assays were used as the standard test for presence of ER and PR until the 1990s. After that, monoclonal antibodies used in IHC analyses became available and quickly replaced ligand‐binding assays because they are easier and cheaper to use and better reflect PR status in the tissue and predict response to hormonal treatment in breast cancer. 52 , 53 Unfortunately, there are still a limited number of studies that related PR‐IHC expression to response to hormonal treatment in EC. Nevertheless, PR positivity was associated with a higher ORR to progestin therapy compared with PR‐negative patients. Among the PR‐negative group, we observed an ORR of just 12.2%, which questions the application of progestin therapy in these patients. Of note, in the study with the highest cutoff value for PR positivity, the highest ORR in PR‐negative tumours was observed. 24 This implies that further research into identification of the optimal cutoff is needed to differentiate between response and no response and improve the predictive value of PR expression. In a recent retrospective study, a novel cutoff value of 50% for PR expression was proposed as all patients with a response had PR expressions greater than 50%. 37 Tumour grade was also related to response to progestin treatment with a higher observed ORR in low‐grade tumours. Tumour grade and ER/PR expression are strongly correlated and ER/PR expression is significantly higher in low‐grade compared with high‐grade tumours. 54 As a result, most responses to hormonal therapy can be found among low‐grade tumours. Nevertheless, a subset of high‐grade tumours have positive PR expression and might therefore be responsive to hormones. 54 In our review, data stratified by both grade and PR status were not reported by individual studies. Further study is indicated to determine the response to progestin treatment in PR‐positive high‐grade ECs.
The incidence of severe toxicity to progestin treatment was low with a comparable risk of thromboembolic processes to treatment with first‐line chemotherapy with carboplatin and paclitaxel. 16 Progestin therapy compares favourably with chemotherapy as severe toxicity occurs in around 35% of chemotherapy‐treated patients. 16
5.3. Strengths and limitations
To our knowledge this is the first systematic review and meta‐analysis that specifically investigates the efficacy and toxicity of the role of PR and grade as predictive biomarkers for progestin therapy in advanced and recurrent EC. There are also some limitations to be addressed. The included studies were mostly retrospective, had moderate or high risk of bias and between‐study heterogeneity was greater than 60% for the main outcome measures. In addition, none of the studies included PR‐IHC data and therefore, available data on the predictive value of PR is solely based on ligand‐binding assays. This shows that high‐quality research into the efficacy of progestin treatment in the population with advanced and recurrent EC is still lacking. The definition for response was defined by 76% (19 of 25 studies reporting ORR) of included studies supporting validity of this outcome measure. However, length of follow up was formalised by just 25% (4 of 16 studies reporting OS or PFS), impacting the validity of OS and PFS outcome measures. There is an urgent need for a prospective study with standardised response criteria, regular follow‐up visits including radiological evaluations and PR‐IHC expression data to study the relation between predictive biomarkers and ORR, PFS and OS.
Similarly, toxicity was defined by a subset of 11 studies and a standardised toxicity scale was applied in 27% (3 of 11) of these studies, although most studies did differentiate between severe and non‐severe toxicity. Grade 3 or 4 toxicity in studies with a standardised toxicity scale did not differ from prevalence of severe toxicity in studies without a standardised toxicity scale.
5.4. Implications for clinical practice and further research
Progestin treatment can be an effective palliative systemic treatment and has the advantage of limited toxicity. Women with advanced and recurrent EC for a large part consist of elderly women with comorbidities so it is essential to have low‐toxicity treatments available. However, the ORR to progestin treatment is still limited and further stratification based on predictive biomarkers is needed. Based on the current study, PR expression appears to be the most promising predictive biomarker for response to progestins. However, more research is needed to optimise the predictive value of PR expression.
The genomic characterisation of EC has increased possibilities to apply targeted treatment to molecular alterations. 55 For example a novel ER pathway activity test that infers the activity of the ER pathway based on the level of mRNAs of established ER target genes has recently been related to progestin therapy in EC. 56 The response rate to progestin therapy in EC with an active ER pathway was 62.1% in this study. 37 In the same study a high correlation between ER pathway activity and PR‐IHC expression was found, suggesting that PR‐IHC expression might be used as a cheap and easy‐to‐use surrogate marker for estrogen‐driven tumour growth. A PR expression cutoff value of more than 50% was suggested in this study as the most relevant cutoff value showing an ORR of 56.8% and a sensitivity of 100%. 37
Genomic characterisation of EC has also resulted in the identification of a novel molecular classification in The Cancer Genome Atlas (TCGA) project. The classification involes three markers (POLE, mismatch repair deficiency, P53 mutation) that are used to classificay cases into four groups with distinct prognosis. There is growing evidence that these markers have not only prognostic but also predictive value for targeted therapy. For progestin therapy however, there is no clear association between TCGA subgroups and response. To date, two studies have found mismatch‐repair status to be potentially predictive for progestin therapy, but the PR status was not incorporated in both studies and the population included atypical hyperplasia and low‐grade, stage I EC instead of advanced stage and recurrent disease. 57 , 58 Therefore, ER and especially PR remain the best investigated biomarkers for the progestin therapy response in this population.
5.5. Conclusion
In this systematic review and meta‐analysis, the efficacy and toxicity of progestin therapy in advanced and recurrent EC is reported. In unselected populations, the ORR to progestin treatment was 30% and the CBR was 52%. PR expression was the most discriminative biomarker with an ORR of 55.4% in PR‐positive EC. The toxicity profile of progestins is favourable. Prospective studies are needed to confirm the predictive value of PR‐IHC expression on response to progestins and investigate the impact on PFS and OS.
AUTHOR CONTRIBUTIONS
Study was conceived and designed by WW and JP. Data were acquired by WW and PB who also performed the quality control of data and algorithms. Data analysis and interpretation were performed by WW, PB, RL, RK, AR and JP, and statistical analysis was performed by WW, PB and JI. The manuscript was prepared by WW and PB, edited and reviewed by WW, PB, RL, JI, RK, AR and JP.
ACKNOWLEDGEMENTS
The authors wish to acknowledge the help of Radboud university medical center to publish this article open access.
FUNDING INFORMATION
There was no funding for the execution of this research.
CONFLICT OF INTERESTS
None declared. Completed disclosure of interests form available to view online as supporting information.
ETHICS STATEMENT
This study was performed in accordance with the declaration of Helsinki. No individual patient data was used, therefore institutional review board was not indicated.
Supporting information
Figure S1
Figure S2
Figure S3
Table S1
Table S2
Table S3
ICMJE
ICMJE
ICMJE
ICMJE
ICMJE
ICMJE
ICMJE
van Weelden WJ, Birkendahl PB, Lalisang RI, IntHout J, Kruitwagen RFPM, Romano A, et al. The effect of progestin therapy in advanced and recurrent endometrial cancer: A systematic review and meta‐analysis. BJOG. 2023;130(2):143–152. 10.1111/1471-0528.17331
Willem Jan van Weelden and Philine B. Birkendahl contributed equally to the manuscript and are joint first authors.
DATA AVAILABILITY STATEMENT
Data used in this review can be made available by the author upon reasonable request.
REFERENCES
- 1. Dunselman GA, Vermeulen N, Becker C, Calhaz‐Jorge C, D'Hooghe T, De Bie B, et al. ESHRE guideline: management of women with endometriosis. Hum Reprod. 2014;29(3):400–12. [DOI] [PubMed] [Google Scholar]
- 2. ACOG Committee on Practice Bulletins–Gynecology . ACOG practice bulletin. Medical management of endometriosis. Number 11, December 1999 (replaces Technical Bulletin Number 184, September 1993). Clinical management guidelines for obstetrician‐gynecologists. Int J Gynaecol Obstet. 2000;71(2):183–96. [DOI] [PubMed] [Google Scholar]
- 3. Heikinheimo O, Fraser I. The current status of hormonal therapies for heavy menstrual bleeding. Best Pract Res Clin Obstet Gynaecol. 2017;40:111–20. [DOI] [PubMed] [Google Scholar]
- 4. Dodd JM, Grivell RM, OBrien CM, Dowswell T, Deussen AR. Prenatal administration of progestogens for preventing spontaneous preterm birth in women with a multiple pregnancy. Cochrane Database Syst Rev. 2019;2019(11):CD012024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Gupta J, Kai J, Middleton L, Pattison H, Gray R, Daniels J, et al. Levonorgestrel intrauterine system versus medical therapy for menorrhagia. N Engl J Med. 2013;368(2):128–37. [DOI] [PubMed] [Google Scholar]
- 6. Group SGOCPECW , Burke WM, Orr J, Leitao M, Salom E, Gehrig P, et al. Endometrial cancer: a review and current management strategies: part II. Gynecol Oncol. 2014;134(2):393–402. [DOI] [PubMed] [Google Scholar]
- 7. Kokka F, Brockbank E, Oram D, Gallagher C, Bryant A. Hormonal therapy in advanced or recurrent endometrial cancer. Cochrane Database Syst Rev. 2010;8(12):CD007926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Lee WL, Yen MS, Chao KC, Yuan CC, Ng HT, Chao HT, et al. Hormone therapy for patients with advanced or recurrent endometrial cancer. J Chin Med Assoc. 2014;77(5):221–6. [DOI] [PubMed] [Google Scholar]
- 9. Aalders JG, Abeler V, Kolstad P. Recurrent adenocarcinoma of the endometrium: a clinical and histopathological study of 379 patients. Gynecol Oncol. 1984;17(1):85–103. [DOI] [PubMed] [Google Scholar]
- 10. Del Carmen MG, Boruta DM 2nd, Schorge JO. Recurrent endometrial cancer. Clin Obstet Gynecol. 2011;54(2):266–77. [DOI] [PubMed] [Google Scholar]
- 11. Creasman WT, Odicino F, Maisonneuve P, Quinn MA, Beller U, Benedet JL, et al. Carcinoma of the corpus uteri. FIGO 26th Annual Report on the Results of Treatment in Gynecological Cancer. Int J Gynaecol Obstet. 2006;95:S105–43. [DOI] [PubMed] [Google Scholar]
- 12. Colombo N, Creutzberg C, Amant F, Bosse T, González‐Martín A, Ledermann J, et al. ESMO‐ESGO‐ESTRO Consensus Conference on Endometrial Cancer: diagnosis, treatment and follow‐up. Int J Gynecol Cancer. 2016. Jan;26(1):2–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Lorusso D, Ferrandina G, Colombo N, Pignata S, Pietragalla A, Sonetto C, et al. Carboplatin‐paclitaxel compared to carboplatin‐paclitaxel‐bevacizumab in advanced or recurrent endometrial cancer: MITO END‐2 ‐ a randomized phase II trial. Gynecol Oncol. 2019;155(3):406–12. [DOI] [PubMed] [Google Scholar]
- 14. Arimoto T, Nakagawa S, Yasugi T, Yoshikawa H, Kawana K, Yano T, et al. Treatment with paclitaxel plus carboplatin, alone or with irradiation, of advanced or recurrent endometrial carcinoma. Gynecol Oncol. 2007;104(1):32–5. [DOI] [PubMed] [Google Scholar]
- 15. Pectasides D, Xiros N, Papaxoinis G, Pectasides E, Sykiotis C, Koumarianou A, et al. Carboplatin and paclitaxel in advanced or metastatic endometrial cancer. Gynecol Oncol. 2008;109(2):250–4. [DOI] [PubMed] [Google Scholar]
- 16. Miller DS, Filiaci VL, Mannel RS, Cohn DE, Matsumoto T, Tewari KS, et al. Carboplatin and paclitaxel for advanced endometrial cancer: final overall survival and adverse event analysis of a phase III trial (NRG Oncology/GOG0209). J Clin Oncol. 2020;38(33):3841–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Thigpen JT, Brady MF, Alvarez RD, Adelson MD, Homesley HD, Manetta A, et al. Oral medroxyprogesterone acetate in the treatment of advanced or recurrent endometrial carcinoma: a dose‐response study by the Gynecologic Oncology Group. J Clin Oncol. 1999;17(6):1736–44. [DOI] [PubMed] [Google Scholar]
- 18. Lentz SS, Brady MF, Major FJ, Reid GC, Soper JT. High‐dose megestrol acetate in advanced or recurrent endometrial carcinoma: a Gynecologic Oncology Group study. J Clin Oncol. 1996;14(2):357–61. [DOI] [PubMed] [Google Scholar]
- 19. Kelley RM, Baker WH. Progestational agents in the treatment of carcinoma of the endometrium. N Engl J Med. 1961;2(264):216–22. [DOI] [PubMed] [Google Scholar]
- 20. van Weelden WJ, Bretveld R, Romano A, van Erp S, Engels S, Lalisang R, et al. Trends over time in the incidence and use of hormonal therapy in endometrial cancer: a population‐based study in the Netherlands. Int J Gynecol Cancer. 2021;31(7):1014–20. [DOI] [PubMed] [Google Scholar]
- 21. Tsai MJ, O'Malley BW. Molecular mechanisms of action of steroid/thyroid receptor superfamily members. Annu Rev Biochem. 1994;63:451–86. [DOI] [PubMed] [Google Scholar]
- 22. Tangen IL, Werner HM, Berg A, Halle MK, Kusonmano K, Trovik J, et al. Loss of progesterone receptor links to high proliferation and increases from primary to metastatic endometrial cancer lesions. Eur J Cancer. 2014;50(17):3003–10. [DOI] [PubMed] [Google Scholar]
- 23. Bartosch C, Monteiro‐Reis S, Vieira R, Pereira A, Rodrigues M, Jeronimo C, et al. Endometrial endometrioid carcinoma metastases show decreased ER‐alpha and PR‐A expression compared to matched primary tumors. PLoS One. 2015;10(8):e0134969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Ehrlich CE, Young PC, Cleary RE. Cytoplasmic progesterone and estradiol receptors in normal, hyperplastic, and carcinomatous endometria: therapeutic implications. Am J Obstet Gynecol. 1981;141(5):539–46. [DOI] [PubMed] [Google Scholar]
- 25. Decruze SB, Green JA. Hormone therapy in advanced and recurrent endometrial cancer: a systematic review. Int J Gynecol Cancer. 2007;17(5):964–78. [DOI] [PubMed] [Google Scholar]
- 26. Lindemann K, Malander S, Christensen RD, Mirza MR, Kristensen GB, Aavall‐Lundqvist E, et al. Examestane in advanced or recurrent endometrial carcinoma: a prospective phase II study by the Nordic Society of Gynecologic Oncology (NSGO). BMC Cancer. 2014;5(14):68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Mileshkin L, Edmondson R, O'Connell RL, Sjoquist KM, Andrews J, Jyothirmayi R, 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(1):29–37. [DOI] [PubMed] [Google Scholar]
- 28. Ethier J‐L, Desautels DN, Amir E, MacKay H. Is hormonal therapy effective in advanced endometrial cancer? A systematic review and meta‐analysis. Gynecol Oncol. 2017;147(1):158–66. [DOI] [PubMed] [Google Scholar]
- 29. Higgins JPT, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta‐analyses. BMJ (Clinical Research Ed). 2003;327(7414):557–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Page MJ, Moher D, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. PRISMA 2020 explanation and elaboration: updated guidance and exemplars for reporting systematic reviews. BMJ. 2021;29(372):n160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Stroup DF, Berlin JA, Morton SC, Olkin I, Williamson GD, Rennie D, et al. Meta‐analysis of observational studies in epidemiology: a proposal for reporting. Meta‐analysis Of Observational Studies in Epidemiology (MOOSE) group. JAMA. 2000. Apr 19;283(15):2008–12. [DOI] [PubMed] [Google Scholar]
- 32. Ouzzani M, Hammady H, Fedorowicz Z, Elmagarmid A. Rayyan‐a web and mobile app for systematic reviews. Syst Rev. 2016;5(1):210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Sterne JACSJ, Page MJ, Elbers RG, Blencowe NS, Boutron I, Cates CJ, et al. RoB 2: a revised tool for assessing risk of bias in randomised trials. BMJ. 2019;366:l4898. [DOI] [PubMed] [Google Scholar]
- 34. Sterne JAC, Reeves BC, Savović J, Berkman ND, Viswanathan M, Henry D, et al. The Risk Of Bias In Non‐randomized Studies – of Interventions (ROBINS‐I) assessment tool. BMJ. 2016;355:i4919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Egger M, Davey Smith G, Schneider M, Minder C. Bias in meta‐analysis detected by a simple, graphical test. BMJ. 1997;315(7109):629–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Schwab M, editor. Broder histological classification. Encyclopedia of Cancer. Berlin, Heidelberg: Springer Berlin Heidelberg; 2011. p. 571. [Google Scholar]
- 37. van Weelden WJ, Lalisang RI, Bulten J, Lindemann K, van Beekhuizen HJ, Trum H, et al. Impact of hormonal biomarkers on response to hormonal therapy in advanced and recurrent endometrial cancer. Am J Obstet Gynecol. 2021;25(4):407.e1–407.e16. [DOI] [PubMed] [Google Scholar]
- 38. Balduzzi S, Rücker G, Schwarzer G. How to perform a meta‐analysis with R: a practical tutorial. Evid Based Ment Health. 2019;22(4):153–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Viechtbauer W. Conducting meta‐analyses in R with the metafor package. J Stat Software. 2010;36(3):1–48. [Google Scholar]
- 40. Sera F, Armstrong B, Blangiardo M, Gasparrini A. An extended mixed‐effects framework for meta‐analysis. Stat Med. 2019;38(29):5429–44. [DOI] [PubMed] [Google Scholar]
- 41. Pandya KJ, Yeap BY, Weiner LM, Krook JE, Erban JK, Schinella RA, et al. Megestrol and tamoxifen in patients with advanced endometrial cancer: an Eastern Cooperative Oncology Group Study (E4882). Am J Clin Oncol. 2001;24(1):43–6. [DOI] [PubMed] [Google Scholar]
- 42. Pautier P, Vergote I, Joly F, Melichar B, Kutarska E, Hall G, et al. A phase 2, randomized, open‐label study of irosustat versus megestrol acetate in advanced endometrial cancer. Int J Gynecol Cancer. 2017;27(2):258–66. [DOI] [PubMed] [Google Scholar]
- 43. Rendina GM, Donadio C, Fabri M, Mazzoni P, Nazzicone P. Tamoxifen and medroxyprogesterone therapy for advanced endometrial carcinoma. Eur J Obstet Gynecol Reprod Biol. 1984;17(4):285–91. [DOI] [PubMed] [Google Scholar]
- 44. Dowdy SC, Mariani A, Bakkum JN, Cliby WA, Keeney GL, Podratz KC. Treatment of pulmonary recurrences in patients with endometrial cancer. Gynecol Oncol. 2007;107(2):242–7. [DOI] [PubMed] [Google Scholar]
- 45. Benraad TJ, Friberg LG, Koenders AJ, Kullander S. Do estrogen and progesterone receptors (E2R and PR) in metastasizing endometrial cancers predict the response to gestagen therapy? Acta Obstet Gynecol Scand. 1980;59(2):155–9. [DOI] [PubMed] [Google Scholar]
- 46. McCarty KS Jr, Barton TK, Fetter BF, Creasman WT, McCarty KS Sr. Correlation of estrogen and progesterone receptors with histologic differentiation in endometrial adenocarcinoma. Am J Pathol. 1979;96(1):171–83. [PMC free article] [PubMed] [Google Scholar]
- 47. Quinn MA, Cauchi M, Fortune D. Endometrial carcinoma: steroid receptors and response to medroxyprogesterone acetate. Gynecol Oncol. 1985;21(3):314–9. [DOI] [PubMed] [Google Scholar]
- 48. Yunokawa M, Yoshida H, Watanabe R, Noguchi E, Shimomura A, Shimoi T, et al. Allred score is a promising predictor of prognosis and medroxyprogesterone acetate efficacy in patients with endometrial cancer. Cancer Chemother Pharmacol. 2017;80(1):127–34. [DOI] [PubMed] [Google Scholar]
- 49. Karagol H, Saip P, Uygun K, Kucucuk S, Aydiner A, Topuz E. Evaluation of prognostic factors and comparison of systemic treatment modalities in patients with recurrent or metastatic endometrial carcinoma. Med Oncol. 2006;23(4):543–8. [DOI] [PubMed] [Google Scholar]
- 50. Piver MS, Barlow JJ, Lurain JR, Blumenson LE. Medroxyprogesterone acetate (Depo‐Provera) vs. hydroxyprogesterone caproate (Delalutin) in women with metastatic endometrial adenocarcinoma. Cancer. 1980;45(2):268–72. [DOI] [PubMed] [Google Scholar]
- 51. Soliman PT, Westin SN, Iglesias DA, Fellman BM, Yuan Y, Zhang Q, et al. Everolimus, letrozole, and metformin in women with advanced or recurrent endometrioid endometrial cancer: a multi‐center, single arm, phase II study. Clin Cancer Res. 2020;26(3):581–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Harvey JM, Clark GM, Osborne CK, Allred DC. Estrogen receptor status by immunohistochemistry is superior to the ligand‐binding assay for predicting response to adjuvant endocrine therapy in breast cancer. J Clin Oncol. 1999;17(5):1474–81. [DOI] [PubMed] [Google Scholar]
- 53. Elledge RM, Green S, Pugh R, Allred DC, Clark GM, Hill J, et al. Estrogen receptor (ER) and progesterone receptor (PgR), by ligand‐binding assay compared with ER, PgR and pS2, by immuno‐histochemistry in predicting response to tamoxifen in metastatic breast cancer: a Southwest Oncology Group Study. Int J Cancer. 2000;89(2):111–7. [PubMed] [Google Scholar]
- 54. van Weelden WJ, Reijnen C, Küsters‐Vandevelde HVN, Bulten J, Bult P, Leung S, 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. [DOI] [PubMed] [Google Scholar]
- 55. Cancer Genome Atlas Research Network , Kandoth C, Schultz N, Cherniack AD, Akbani R, Liu Y, et al. Integrated genomic characterization of endometrial carcinoma. Nature. 2013;497(7447):67–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. van Weelden WJ, van der Putten LJM, Inda MA, van Brussel A, Snijders M, Schriever LMM, et al. Oestrogen receptor pathway activity is associated with outcome in endometrial cancer. Br J Cancer. 2020;123(5):785–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Raffone A, Catena U, Travaglino A, Masciullo V, Spadola S, Della Corte L, et al. Mismatch repair‐deficiency specifically predicts recurrence of atypical endometrial hyperplasia and early endometrial carcinoma after conservative treatment: a multi‐center study. Gynecol Oncol. 2021;161(3):795–801. [DOI] [PubMed] [Google Scholar]
- 58. Zakhour M, Cohen JG, Gibson A, Walts AE, Karimian B, Baltayan A, et al. Abnormal mismatch repair and other clinicopathologic predictors of poor response to progestin treatment in young women with endometrial complex atypical hyperplasia and well‐differentiated endometrial adenocarcinoma: a consecutive case series. BJOG. 2017;124(10):1576–83. [DOI] [PubMed] [Google Scholar]
- 59. Anderson DG. Management of advanced endometrial adenocarcinoma with medroxyprogesterone acetate. Am J Obstet Gynecol. 1965;92:87–99. [DOI] [PubMed] [Google Scholar]
- 60. Malkasian GD Jr, Decker DG, Mussey E, Johnson CE. Progestogen treatment of recurrent endometrial carcinoma. Am J Obstet Gynecol. 1971;110(1):15–23. [DOI] [PubMed] [Google Scholar]
- 61. Morrison J, Blunt VA. Palliation of metastatic endometrial adenocarcinoma with dimethisterone. Aust N Z J Obstet Gynaecol. 1972;12(1):27–9. [DOI] [PubMed] [Google Scholar]
- 62. Rozier JC Jr, Underwood PB Jr. Use of progestational agents in endometrial adenocarcinoma. Obstet Gynecol. 1974;44(1):60–4. [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Figure S1
Figure S2
Figure S3
Table S1
Table S2
Table S3
ICMJE
ICMJE
ICMJE
ICMJE
ICMJE
ICMJE
ICMJE
Data Availability Statement
Data used in this review can be made available by the author upon reasonable request.
